WO2017219916A1 - Molecule, cell expressing the same, and preparation method and use thereof - Google Patents

Molecule, cell expressing the same, and preparation method and use thereof Download PDF

Info

Publication number
WO2017219916A1
WO2017219916A1 PCT/CN2017/088594 CN2017088594W WO2017219916A1 WO 2017219916 A1 WO2017219916 A1 WO 2017219916A1 CN 2017088594 W CN2017088594 W CN 2017088594W WO 2017219916 A1 WO2017219916 A1 WO 2017219916A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
ctl
tumor
domain
receptor molecule
Prior art date
Application number
PCT/CN2017/088594
Other languages
French (fr)
Chinese (zh)
Inventor
李鹏
魏新茹
叶未
赵若聪
赖允鑫
林思妙
姚瑶
Original Assignee
深圳市体内生物医药科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 深圳市体内生物医药科技有限公司 filed Critical 深圳市体内生物医药科技有限公司
Priority to CN202110620458.7A priority Critical patent/CN113512126B/en
Priority to CN201780000466.3A priority patent/CN107624119B/en
Publication of WO2017219916A1 publication Critical patent/WO2017219916A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70596Molecules with a "CD"-designation not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian

Definitions

  • the present invention relates to the field of cellular immunotherapy technology for tumors, and in particular to a chimeric immunosuppressive checkpoint receptor molecule, a nucleic acid encoding the same, a construct containing the nucleic acid, an expression vector and transformed cells, and Pharmaceutical use.
  • the tumor microenvironment can down-regulate the number of tumor antigen-specific helper/toxic killer cells and the release of inflammatory factors, and up-regulate the number and inhibitory functions of immunosuppressive cells, such as regulatory T cells and myeloid-derived suppressor cells ( Myeloid-Derived Suppressor Cells) subvert the normal immune system as a dominant factor in promoting tumor growth.
  • immunosuppressive cells such as regulatory T cells and myeloid-derived suppressor cells ( Myeloid-Derived Suppressor Cells) subvert the normal immune system as a dominant factor in promoting tumor growth.
  • a blocking inhibition signaling pathway to down-regulate tumor microscopy.
  • a method for inhibiting cells in the environment such as specifically targeting PD-1 (Programmed Death 1), PD-L1 (Programmed Death Ligand 1), CTLA4 (Cytotoxic T-Lymphocyte-associated Antigen-4) antibody or inhibitor, Immune activators and the like.
  • the application of the immunosuppressive-associated antigens PD-1 and CTLA-4 to tumor immunotherapy is usually to construct a PD-1 antibody (Pembrolizumab, Nivolumab) or a CTLA-4 antibody (Ipilimumab), or use PD- 1 or an inhibitor of CTLA-4 or a signaling pathway blocker.
  • PD-1 antibodies and CTLA-4 antibodies have certain limitations, that is, antibodies need to penetrate into tumor tissues to exert therapeutic effects, so the clinical application of antibodies is uncertain. It has been reported that when CAR-GD2T cells are combined with PD-1 antibody for immunotherapy, they can resist the immunosuppressive effect of the tumor microenvironment and enhance the tumor killing of CAR T cells.
  • CAR T and inhibitory CAR T co-expression of targeting tumor-associated antigens CAR T and inhibitory CAR T has also been reported in the prior art (the extracellular antigen is a normal cell antigen and the intracellular domain is PD-1). Or the inhibitory signal domain of CTLA-4) prevents the occurrence of CAR T off-target (off-target phenomenon: CAR T cells recognize and kill normal cells).
  • CAR T co-expressing the diabody PD-1 and CD19/Mesothelin/PSCA has also been reported in the prior art, although this expands the target range (expressing PD-1 or CD19/Mesothelin/PSCA or both) Tumor cells), but neglecting the side effects caused by blindly increasing cell killing: cytokine storm, off-target phenomenon (normal cells also express two antigens or one, causing CAR T to kill normal cells, causing functional defects in normal cell loss) ).
  • the transformed cell expressing the chimeric immunosuppressive checkpoint receptor molecule of the present invention can create a tumor immunosuppressive microenvironment with enhanced tumor killing effect; and the transformed cell has self-regulating function and can be alleviated Excessive side effects such as non-specific immune effects and cytokine storms.
  • the invention provides a chimeric immunosuppressive checkpoint receptor molecule comprising an extracellular domain, a transmembrane domain and an intracellular domain, wherein the extracellular domain and optionally The transmembrane domain is the corresponding domain of an immunosuppressive checkpoint receptor molecule or is genetically engineered based on the domain, the intracellular domain being one or more immune activation signal domains.
  • the immunosuppressive checkpoint receptor molecule is selected from the group consisting of PD-1, CTLA-4, LAG3, TIM3, A2AR, B7H3, B7H4, BTLA, IDO, KIR, CD160, 2B4 (CD244) and VISTA (C10orf54);
  • the immune checkpoint receptor molecule is PD-1, CTLA-4, LAG3 or TIM3.
  • the intracellular domain comprises an immunostimulatory signal combination and an optional signal peptide
  • said immune costimulatory signal combination comprises a TLR1 and/or TLR2 signal domain
  • the intracellular domain is a combination of TLR1 and/or TLR2 and a 41BB and/or CD28 signaling domain.
  • the optional signal peptide is a signal peptide corresponding to an immunosuppressive checkpoint receptor molecule.
  • the intracellular domain further comprises a CD3 sputum intracellular domain
  • the TLR1 and/or TLR2 signal domain and optionally the 41BB and/or CD28 signal domain are arranged on the N-terminal side of the CD3 cell intracellular domain.
  • the invention provides a nucleic acid encoding a chimeric immunosuppressive checkpoint receptor molecule of the first aspect.
  • the present invention provides a nucleic acid construct comprising the nucleic acid of the second aspect, and an immunosuppressive checkpoint receptor molecule operably linked thereto for directing said chimerism in a host cell One or more control sequences expressed.
  • the present invention provides an expression vector comprising the nucleic acid construct of the third aspect; preferably, the expression vector is a lentiviral expression vector.
  • the present invention provides a transformed cell comprising the nucleic acid of the second aspect, or wherein the nucleic acid construct of the third aspect or the expression vector of the fourth aspect is transformed;
  • the transformed cell is an immune cell, further preferably a T cell, a B cell or an NK cell.
  • the transformed cell is a Zeus sulphate cytotoxic T lymphocyte (S-CTL), which is a lymphocyte that overexpresses a checkpoint molecule that undergoes immunosuppression to activation, particularly T lymphocytes.
  • S-CTL cells include SP-CTL cells expressing a chimeric PD-1 receptor molecule, SC-CTL cells expressing a chimeric CTLA-4 receptor molecule, and ST-CTL cells expressing a chimeric TIM3 receptor molecule. And SL-CTL cells expressing a chimeric LAG3 receptor molecule and the like. S-CTL cells obtain lymphocytes resistant to immunosuppression by transfecting a chimeric immunosuppressive-agonistic transition checkpoint molecule gene into human T cells.
  • S-CTL lymphocytes have the following characteristics:
  • S-CTL contains natural immunosuppressive checkpoint receptor molecules (such as PD-1, CTLA-4, TIM3, LAG3, etc.) expressed on the surface of immune effector cells, especially T cells, NK cells, etc., and tumor cells or tumors.
  • immune activation signal downstream of the S-CTL vector can be activated to promote the activation, proliferation, recognition, killing and other functions of the immune cell itself;
  • S-CTL structure extracellular segment and transmembrane region are completely natural immunosuppressive checkpoint molecular structures for specific recognition of tumor cell surface ligands and recognition signals for efficient delivery in S-CTL cells; intracellular The region is one or more immune activation signal domains, wherein the present invention finds an intracellular domain comprising a TLR1 and/or TLR2 signaling activation domain, which further promotes tumor killing effects;
  • the invention provides a method of producing a transformed cell of the fifth aspect, comprising: introducing a nucleic acid of the second aspect into a cell, or transforming the nucleic acid construct of the third aspect Or the step of expressing the vector as described in the fourth aspect.
  • the invention provides a chimeric immunosuppressive checkpoint receptor molecule according to the first aspect, a nucleic acid according to the second aspect, a nucleic acid construct according to the third aspect, the fourth aspect
  • the expression vector or the transformed cell of the fifth aspect in the preparation of a ligand corresponding to an immunosuppressive checkpoint receptor molecule, preferably with PD-L1, B7.1, B7.2, Galectin-9, MHC Use of a drug for expression of a disease associated with II;
  • the disease is a tumor corresponding to a ligand expressing an immunosuppressive checkpoint receptor molecule, preferably a tumor expressing PD-L1, B7.1, B7.2, Galectin-9, MHC II;
  • the tumor corresponding to the immunosuppressive checkpoint receptor molecule is selected from the group consisting of lung cancer, leukemia, breast cancer, Prostate cancer, pancreatic cancer, liver cancer, melanoma, and non-melanoma skin cancer.
  • an "Immune Checkpoint” is a molecule that up-regulates or down-regulates an immunostimulatory signal in the immune system, and is classified into a Stimulatory Checkpoint and an Inhibitory Checkpoint molecule. ).
  • An “immunosuppressive checkpoint” is a molecule that down-regulates an immune stimulus signal in the immune system, such as: PD-1, CTLA-4, TIM3, LAG3, A2AR, B7-H3, B7-H4, BTLA, IDO, KIR, CD160, 2B4 (CD244), VISTA (C10orf54), TIGIT, LAIR1, etc.
  • Immunoactivation signal During lymphocyte activation, T cells need to acquire two signals for full activation; the first signal is antigen-specific, that is, through the T cell receptor and the antigen-presenting cell APC surface MHC molecule junction The second signal, the costimulatory signal, is antigen-unspecific and is produced by the binding of T cells and costimulatory signaling molecules on the surface of APC cells.
  • the intracellular domain of the chimeric immunosuppressive checkpoint receptor molecule of the invention may comprise costimulatory signaling molecules CD28, OX40, ICOS (CD278), 4-1BB (CD137), CD40, CD27, CD134, CDS, ICAM- 1.
  • the transformed cell of the present invention is a novel tumor killer cell which specifically recognizes cells expressing an immunosuppressive checkpoint molecular ligand in a tumor suppressing immunosuppressive microenvironment, has a low off-target rate; and, its intracellular domain
  • the unique design can further promote its tumor killing effect, and can effectively eliminate the tumor immunosuppressive effect;
  • the immune effector cells (such as T cells, etc.) transformed by the present invention are induced to express a certain amount of natural immunosuppression in the tumor microenvironment.
  • the activated vector transformed by the present invention is optimized for immune activation, and comprises an intrinsic immune activation signal TLR1/TLR2 intracellular segment, and a combination of 4-1BB, CD28 and other immune activation molecules to construct a co-stimulatory signal, and finally a CD3 ⁇ chain
  • S-CTL cells have strong functions of killing tumor cells and tumor-associated immunosuppressive cells.
  • SC-CTL cells can specifically recognize the microenvironment in killing tumors.
  • CD80+/CD86+ tumor cells and tumor-associated immunosuppressive cells synergistically promote anti-tumor immune effects.
  • the transformed cells expressing the chimeric receptor molecule of the present invention have a good clinical application prospect in antitumor therapy.
  • Figure 1 shows the synthetic sequence map in Example 1.
  • Figure 2 shows the pWPXLd-S(PD1)-CTL-2A-EGFP plasmid constructed in Example 1, pWPXLd-S(CTLA-4)-CTL-2A-EGFP plasmid, pWPXLd-S(TIM3)-CTL-2A- EGFP plasmid and pWPXLd-S (LAG3)-CTL-2A-EGFP plasmid map.
  • Figure 3 shows the positive rate of viral infection of SP-CTL cells by flow cytometry; blank group: non-transfected T cells stained without CD3 flow cytometry.
  • Figure 4 shows the positive rate of viral infection of SC-CTL cells by flow cytometry; normal T cell group: non-transfected T cells.
  • Figure 5 shows the killing effect of SP-CTL cells on the human T-ALL cell line JURKAT-GL.
  • Figure 6 shows the killing effect of SP-CTL cells on human non-small cell lung cancer H460-GL.
  • Figure 7 shows the killing effect of SC-CTL cells on the human B-ALL cell line NALM6-GL.
  • Figure 8 shows the in vivo killing effect of SP-CTL cells on human non-small cell lung cancer H460-GL.
  • Figure 9 shows the in vitro killing effect of SP-CTL cells on A549 cells that do not express PD-L1.
  • Figure 10 shows the in vitro killing effect of SP-CTL cells on K562 cells that do not express PD-L1.
  • Figure 11 is a graph showing the results of flow cytometry detection of PD-L1 expression in lung cancer cell lines H1299, H460, H23 and A549 and prostate cancer cell line PC3.
  • Figure 12 shows the in vitro killing effect of SP-CTL cells on PD-L1 expressing lung cancer cell lines H1299, H460, H23 and A549 and prostate cancer cell line PC3.
  • Figure 13 is a graph showing the results of in vivo recognition and killing of primary lung cancer cells by SP-CTL cells; wherein Fig. 13-A shows the average volume results of tumor tissue blocks in each experimental group at different time points, and Fig. 13-B shows each experimental group. The size of the three replicated tumor tissue blocks is aligned with the picture.
  • Figure 14 is a graph showing the results of in vivo recognition and killing of primary gastric cancer cells by SP-CTL cells; wherein Figure 14-A shows the size comparison of three replicate tumor tissue blocks in each experimental group, and Figure 14-B shows the experimental groups. The average weight result of the tumor tissue mass.
  • Figure 15 is a graph showing the results of in vivo recognition and killing of primary osteosarcoma cells by SP-CTL cells; wherein Figure 15-A shows a size comparison of six replicate tumor tissue blocks in each experimental group, and Figure 15-B shows each experiment. The average weight result of the group of tumor tissue blocks.
  • Figure 16 is a graph showing the results of in vitro specific recognition and killing of non-Hawsonson lymphoma RL cells by SC-CTL cells.
  • Figure 17 is a graph showing the results of in vivo recognition and killing of non-Hawking's lymphoma RL cells by SC-CTL cells;
  • Figure 17-A shows a comparison of the size of two replicate tumor tissue blocks in each experimental group,
  • Figure 17- B shows the average weight result of the tumor tissue mass of each experimental group.
  • Figure 18 is a graph showing the results of in vivo recognition and killing of primary osteosarcoma cells by mSC-CTL cells; wherein Figure 18-A shows the size comparison of the tumor tissue blocks of each experimental group, and Figure 18-B shows the experimental groups. The average weight result of the tumor tissue mass.
  • Figure 19 is a graph showing the results of SP-CTL, mSC-CTL alone or in combination to specifically recognize and kill primary lung cancer cells in vivo;
  • Figure 19-A shows the average volume results of tumor tissue blocks in each experimental group,
  • Figure 19-B The average weight results of the tumor tissue blocks of each experimental group are shown.
  • Figure 20 shows that SP-CTL, mSC-CTL alone or in combination with CD80+ cells (Fig. 20-A), CD86+ cells (Fig. 20-B) and CD11b+Ly6G+ cells (MDSC) in tumor tissues in vivo (Fig. 20- C) The impact.
  • Figure 21 shows that the B acute lymphoblastic leukemia cell line Nalm6, RL, Raji highly expresses the Lag3 ligand MHC II.
  • Figure 22 shows that SL-CTL cells specifically recognize and kill the B-ALL cell line Nalm6 cells that highly express MHC II ligand.
  • Figure 23 shows that SL-CTL cells co-incubated with tumor cells significantly up-regulated protein expression and secretion of inflammatory factors IL-2 (Figure 23A), IFN- ⁇ ( Figure 23A).
  • Figure 24 shows that A549 cells do not express Galectin9 (Tim3 ligand) surface protein in vitro, but in vivo micro
  • the environment-induced lung cancer cell line A549 specifically expresses Galectin9 surface protein.
  • Figure 25 shows that primary lung cancer cells 1, 2, and 3 specifically express Galectin9 surface protein in a patient-derived tumor xenograft (PDX) model.
  • PDX patient-derived tumor xenograft
  • Figure 26 shows that ST-CTL specifically recognizes primary lung cancer cells that kill high expression of Galectin9 surface protein in the PDX model.
  • immunostimulatory checkpoint molecule signal peptide + extracellular segment + transmembrane region
  • immunostimulatory signal 1 intracellular segment
  • immunostimulatory signal 2 generally TLR1 or TLR2
  • intracellular segment - CD3 The DNA sequence, the first and the end of the DNA sequence contain the PmeI and Spe1 restriction sites, respectively.
  • the synthetic sequence map is shown in Figure 1.
  • SEQ NO. 1 for the preparation of S-CTL targeting PD-1, ie, SP-CTL, the synthesized DNA sequence, the structure is PD-1 signal peptide + extracellular Segment + transmembrane region - CD28-TLR1/2-CD3 ⁇
  • SEQ NO. 2 for the preparation of S-CTL targeting CTLA-4, ie, SC-CTL, the synthesized DNA sequence, the structure is CTLA-4 signal peptide + extracellular segment + transmembrane region - CD28 intracellular segment - TLR1/2 intracellular segment - CD3 ⁇
  • SEQ NO. 2 for the preparation of S-CTL targeting CTLA-4, ie, SC-CTL, the synthesized DNA sequence, the structure is CTLA-4 signal peptide + extracellular segment + transmembrane region - CD28 intracellular segment - TLR1/2 intracellular segment - CD3 ⁇
  • S-CTL Zinc Cytotoxic T Lymphocyte
  • S-CTL chimera Receptors are T lymphocytes that enhance immune killing ability, and are not limited to T cells, and immune effector cells overexpressing S-CTL chimeric receptors also have enhanced immunological killing function.
  • S-CLT chimeric receptors ie chimeric immunosuppressive checkpoint molecules, extracellular and transmembrane segments are natural human immunosuppressive checkpoint receptor molecules such as PD-1, CTLA-4, TIM3, LAG3, A2AR , B7-H3, B7-H4, BTLA, IDO, KIR, CD160, 2B4 (CD244), VISTA (C10orf54), TIGIT, LAIR1, etc.
  • the intracellular segment is an immunostimulatory signaling molecule comprising TLR1/TLR2 Combination with intracellular segments such as TLR3-10, 4-1BB, CD28, ICOS, CD27, CD2, OX40, CD30, CD40, LFA-1, CD7, CD2S, LIGHT, NKG2C, B7-H3, CD83, etc.
  • the CD3zeta chain is composed of a combination; it can specifically recognize and kill tumor cells and tumor-associated immunosuppressive cells expressing immunosuppressive checkpoint ligands.
  • SP-CTL an S-CTL cell expressing a chimeric PD-1 receptor, specifically recognizes tumor cells and tumor-associated stromal cells or immunosuppressive cells that secrete PD-L1.
  • SC-CTL a S-CTL cell expressing a chimeric CTLA-4 receptor, specifically recognizes tumor cells and tumor-associated stromal cells or immunosuppressive cells that kill B7.1 and/or B7.2.
  • ST-CTL an S-CTL cell expressing a chimeric TIM3 receptor, specifically recognizes tumor cells and tumor-associated stromal cells or immunosuppressive cells that secrete Galectin-9.
  • SL-CTL a S-CTL cell expressing a chimeric LAG3 receptor, specifically recognizes tumor cells that secrete MHC II and tumor-associated stromal cells or immunosuppressive cells.
  • pWPXLd-S(PD1/CTLA-4/TIM3/LAG3)-CTL-2A-EGFP plasmid was obtained with pWPXLd-2A-EGFP, linearized SEQ NO.2 and pWPXLd-2A-EGFP, and the plasmid map is shown in the attached drawing. 2.
  • the pWPXLd-S (PD1/CTLA-4/TIM3/LAG3)-CTL-2A-EGFP plasmid was transfected into the 293T together with the lentiviral helper plasmid (pMD2.G, psPAX2) with PEI.
  • the cells were co-transfected with the empty vector pWPXLd-EGFP and the helper plasmid as a control group;
  • T cells were sorted by MACS Pan-T magnetic beads and diluted to a concentration of 2.5 ⁇ 10 6 with T cell medium (AIM-V plus 5% FBS, penicillin 100 U/ml and streptomycin 0.1 mg/ml). /ml to be used;
  • Example 2 In vitro killing effect of SP-CTL cells on human leukemia cell line JURKAT
  • the RLU relative light unit
  • Formula for killing ratio 100% ⁇ (blank hole reading - experimental hole reading) / blank hole reading, the in vitro killing ratio of SP-CTL cells to human leukemia cell line JURKAT is shown in FIG. 5 .
  • Example 3 In vitro killing effect of SP-CTL cells on non-small cell lung cancer H460
  • the RLU relative light unit
  • Example 4 In vitro killing effect of SC-CTL cells on human leukemia cell line NALM6
  • SC-CTL virus-infected mixed cells with 1 ⁇ 10 of GFP-positive cells were 3.5 ⁇ 10 4 , 1.75 ⁇ 10 4 , 8.6 ⁇ 10 3 , 4.4 ⁇ 10 3 , 2.2 ⁇ 10 3 , and 1.1 ⁇ 10 3 , respectively .
  • Four NALM6-GL cells were co-cultured (the control group was GFP T cells, the blank group was no T cells, and each group was set with three duplicate wells);
  • the RLU relative light unit
  • Killing ratio calculation formula 100% ⁇ (blank hole reading - experimental hole reading) / blank hole reading, the in vitro killing ratio of SC-CTL cells to human leukemia cell line NALM6 is shown in FIG. 7 .
  • Example 5 In vivo killing effect of SP-CTL cells on human non-small cell lung cancer H460
  • the SP-CTL cells of the present invention have significant in vivo and exogenous killing effects on various tumors expressing the corresponding ligands.
  • SP-CTL cells have a specific killing effect only on target cells expressing PD-L1, no killing effect on cells not expressing PD-L1, and have stable biosafety.
  • SP-CTL GFP-positive cells were separately labeled with H1299, H460, H23, A549, and PC3 cells labeled with luciferase GL (H1299-GL, H460-GL, H23-GL, A549-GL, PC3-GL, respectively)
  • the cells were co-cultured in a ratio of 1:1, 1:2, 1:4, 1:8, 1:16 (the control group was GFP T cells, and three replicate wells were set in each group);
  • SP-CTL cells can specifically recognize and kill target cells (lung cancer cells, prostate cancer cells, etc.) expressing PD-L1.
  • Tumor tissue blocks were obtained from the mouse model of lung cancer on the 44th day after tumor transplantation; the size alignment of the three replicated tumor tissue blocks in each experimental group is shown in Fig. 13-B.
  • the SP-CTL cells of the present invention can effectively eliminate primary lung cancer cells in vivo in the case of primary tumor heterogeneity and immunosuppressive microenvironment.
  • Example 9 SP-CTL cells were identified in vivo to kill primary gastric cancer cells
  • SP-CTL virus-infected mixed cell with SP-CTL GFP positive cells of 4 ⁇ 10 6 was injected into a mouse model of gastric cancer, and a second injection of 4 ⁇ 10 6 SP-CTL virus infection was mixed for 25 days.
  • the cells and the control group were tumor-bearing mice injected with GFP T cells, and three replicates were set for each experimental group;
  • the SP-CTL cells of the present invention can effectively eliminate primary gastric cancer cells in vivo in the case of primary tumor heterogeneity and immunosuppressive microenvironment.
  • Example 10 SP-CTL cells in vivo to identify primary osteosarcoma cells
  • the SP-CTL cells of the present invention can effectively remove primary osteosarcoma cells in vivo in the case of primary tumor heterogeneity and immunosuppressive microenvironment.
  • SC-CTL cells can specifically recognize and kill non-Hawkinson lymphoma cells in vitro.
  • Example 12 SC-CTL cells specifically recognize killer non-Hawking's lymphoma RL cells in vivo
  • tumor tissue blocks were obtained from the mouse model of RL-GL tumor-bearing mice; the size comparison of two repeated tumor tissue blocks in each experimental group is shown in Figure 17-A. Tumor tissue of each experimental group The average weight result of the block is shown in Figure 17-B.
  • SC-CTL cells of the present invention can effectively eliminate non-Hawsonson lymphoma cells in vivo.
  • Example 13 mSC-CTL cells specifically recognize killer primary osteosarcoma cells in vivo
  • Tumor tissue blocks were obtained from the osteosarcoma mouse model 44 days after tumor transplantation; the size of the tumor tissue blocks in each experimental group was shown in Fig. 18-A, and the tumor tissue blocks of each experimental group were The average weight results are shown in Figure 18-B.
  • the mSC-CTL cells of the present invention can effectively eliminate primary osteosarcoma cells in vivo in the case of primary tumor heterogeneity and immunosuppressive microenvironment.
  • Example 14 in combination with mSC-CTL and SP-CTL to identify killing tumor cells in vivo
  • mouse extracellular domain + transmembrane region (nucleotide sequence and amino acid sequence as shown in SEQ. NO 5, 6) by mouse cloning means (h) extracellular domain + transmembrane region of SC-CTL , obtaining mSC-CTL; the purpose of this example is to evaluate the scavenging effect of SP-CTL and mSC-CTL on tumor cells, and evaluate SC-CTL recognizes the killing effect of mouse CD80+/CD86+ tumor helper cells (non-tumor cells) and evaluates the effect of this effect on tumor clearance.
  • the specific procedures are as follows:
  • the lung cancer mouse model was divided into four groups, and SP-CTL virus-infected mixed cells (SP-CTL group) with SP-CTL GFP positive cells of 2 ⁇ 10 5 were injected, and mSC-CTL GFP-positive cells were injected.
  • SP-CTL group SP-CTL virus-infected mixed cells
  • mSC-CTL group 2 ⁇ 10 5 SP-CTL virus-infected mixed cells
  • SP-CTL+ 2 ⁇ 10 5 SP-CTL GFP-positive cells and 2 ⁇ 10 5 mSC-CTL GFP-positive cells
  • mSC-CTL can not only identify killing tumor cells, but also recognize CD80+ or/and CD86+ cells (except tumor helper cells other than tumor cells), and mSC-CTL for CD80+/CD86+
  • the killing effect of tumor cells can effectively inhibit tumor growth.
  • mSC-CTL combined with SP-CTL can simultaneously identify killer tumor cells and CD80+/CD86+ tumor-assisted cells, and block the effects of CD80+/CD86+ tumor-assisted cells on tumor cell growth, migration, drug resistance, immune escape, etc. Tumor elimination effect fruit.
  • HLA-DR is a key component of MHC II molecule.
  • the lines all specifically express human HLA-DR gene, which specifically expresses MHC II molecules (Lag3 ligand), indicating that human B cell leukemia and B cell lymphoma tumor cells are potential target cells of SL-CTL.
  • Example 16 SL-CTL specifically recognizes tumor cells that secrete Lag3 ligand ( Figures 22, 23)
  • SL-CTL cells were obtained by packaging (p. 1) lentivirus pWPXLd-SL-CTL-2A-EGFP plasmid and infecting human primary T cells.
  • SL-CTL cells (blank control group GFP-T) and Nalm6-GL cells were co-cultured for 1:1 with 1:1, and the interleukin-2 (IL-2) and interference in the culture supernatant were detected by ELISA.
  • the level of gamma (IFN ⁇ ) is shown in Figure 23. The results showed that SL-CTL cells were able to kill Nalm6 target cells more efficiently than wild T cells (GFP-T group), and SL-CTL cells secreted more inflammatory factors IL-2 and IFN ⁇ .
  • Example 17 tumor cells in vivo can up-regulate the expression of its own TIM3 ligand Galectin9 ( Figures 24, 25).
  • A549 cells were transplanted into immunodeficient mice (such as NOD/SCID IL2rg-/-), 21 days later, The tumor block of the mouse was taken, ground and lysed into a single cell suspension, and the proportion of Galectin9-positive cells was detected, as shown in FIG. 24;
  • tumor cells in vivo (closer to the tumor state in the patient) specifically up-regulate the expression of Galectin9 (Tim ligand) relative to the in vitro cultured tumor cell line, indicating in vivo status (including patients)
  • Tumor cells of tumor cells in an in vivo state are potential specific target cells of ST-CTL cells.
  • tumor tissue blocks were obtained from three groups of experimental lung cancer mouse models; the tumor tissue mass was weighed, and the results are shown in Fig. 26.
  • ST-CTL cells can inhibit the growth of tumor cells in vivo.

Abstract

Provided are a chimeric immune inhibitory checkpoint receptor molecule, a nucleic acid encoding the same, a construct, expression vector and transformed cell containing the nucleic acid, and a pharmaceutical use thereof. The chimeric immune inhibitory checkpoint receptor molecule comprises an extracellular domain, a transmembrane domain, and an intracellular domain. The extracellular domain and the optional transmembrane domain are corresponding domains of an immune inhibitory checkpoint receptor molecule or are genetically engineered on the basis of the corresponding domains. The intracellular domain is one or more immune activation signal domains. The transformed cell has a strong tumor-killing effect and can abrogate a tumor-immunosuppressive microenvironment, thereby strengthening an immune response in the tumor microenvironment. The transformed cell also has a self-regulation function and can alleviate side effects such as an overactive, non-specific immune response and a cytokine storm.

Description

一种分子、表达其的细胞及其制备方法和用途Molecule, cell expressing the same, preparation method and use thereof 技术领域Technical field
本发明涉及肿瘤的细胞免疫治疗技术领域,具体地,涉及一种嵌合的免疫抑制检查点受体分子,编码其的核酸,含有该核酸的构建体、表达载体和转化的细胞,以及它们的制药用途。The present invention relates to the field of cellular immunotherapy technology for tumors, and in particular to a chimeric immunosuppressive checkpoint receptor molecule, a nucleic acid encoding the same, a construct containing the nucleic acid, an expression vector and transformed cells, and Pharmaceutical use.
背景技术Background technique
CAR-T细胞肿瘤治疗现状Current status of CAR-T cell tumor therapy
2015年10月,国家癌症中心在著名癌症专业期刊《Cancer Letters》上首次发布我国居民癌症现患数据。结果显示,我国5年内诊断为癌症且仍存活的病例数约为749万(其中男性患者368万人,女性患者381万人),总体5年癌症患病率为556/10万,数据从整体上反映了癌症病人对新治疗研究进展的迫切需求。In October 2015, the National Cancer Center released the cancer data of residents in China for the first time in the famous cancer professional journal "Cancer Letters". The results showed that the number of cases diagnosed as cancer in China within 5 years and still survivable was about 7.49 million (including 3.86 million male patients and 3.81 million female patients). The overall 5-year cancer prevalence rate was 556/100,000. It reflects the urgent need for cancer patients to progress in new treatment research.
而免疫疗法已然成为人类对抗癌症新的希望,而CAR-T治疗血液肿瘤特别是B-ALL白血病的临床试验结果,使其成为了近年来最备受瞩目的肿瘤免疫疗法之一。然而靶向实体瘤的CAR T免疫治疗研究仍然面对巨大的考验:如何突破实体瘤组织壁垒、如何抵抗肿瘤组织内的免疫抑制、免疫细胞如何在缺氧微环境中竞争生存等。Immunotherapy has become a new hope for humans to fight cancer, and the clinical trial results of CAR-T in the treatment of hematological tumors, especially B-ALL leukemia, have made it one of the most well-received tumor immunotherapy in recent years. However, the CAR T immunotherapy research targeting solid tumors still faces a huge test: how to break through the solid tumor tissue barrier, how to resist the immunosuppression in the tumor tissue, and how the immune cells compete for survival in the hypoxic microenvironment.
肿瘤治疗的难题,特别是实体瘤及其肿瘤微环境中的免疫抑制作用The problem of tumor treatment, especially the immunosuppression in solid tumors and their tumor microenvironment
肿瘤微环境可下调肿瘤抗原特异性的辅助/毒性杀伤细胞的数量和炎症因子的释放,上调免疫抑制细胞数量和抑制功能,如调节性T细胞(Regulatory T cells)和髓系来源的抑制细胞(Myeloid-Derived Suppressor Cells),将正常免疫系统颠覆为促进肿瘤自身生长的优势因素。The tumor microenvironment can down-regulate the number of tumor antigen-specific helper/toxic killer cells and the release of inflammatory factors, and up-regulate the number and inhibitory functions of immunosuppressive cells, such as regulatory T cells and myeloid-derived suppressor cells ( Myeloid-Derived Suppressor Cells) subvert the normal immune system as a dominant factor in promoting tumor growth.
针对肿瘤免疫抑制的相应措施现状Status of corresponding measures against tumor immunosuppression
针对实体瘤免疫抑制的情况,科学家陆续研发出阻断抑制信号通路,下调肿瘤微 环境中的抑制性细胞的方法,如特异性靶向PD-1(Programmed Death 1)、PD-L1(Programmed Death Ligand 1)、CTLA4(Cytotoxic T-Lymphocyte-associated Antigen-4)抗体或抑制剂,免疫激活因子等。In response to the immunosuppression of solid tumors, scientists have successively developed a blocking inhibition signaling pathway to down-regulate tumor microscopy. A method for inhibiting cells in the environment, such as specifically targeting PD-1 (Programmed Death 1), PD-L1 (Programmed Death Ligand 1), CTLA4 (Cytotoxic T-Lymphocyte-associated Antigen-4) antibody or inhibitor, Immune activators and the like.
在现有技术中,将免疫抑制相关抗原PD-1和CTLA-4应用于肿瘤免疫治疗的手段通常是构建PD-1抗体(Pembrolizumab、Nivolumab)或CTLA-4抗体(Ipilimumab),或者使用PD-1或CTLA-4的抑制剂或信号通路阻断剂。然而,PD-1抗体和CTLA-4抗体存在一定的限制性,即,抗体需要渗入肿瘤组织中才可发挥疗效,所以抗体的临床应用具有不确定性。据报道,CAR-GD2T细胞联合PD-1抗体进行免疫治疗时,可抵抗肿瘤微环境的免疫抑制作用,加强CAR T细胞肿瘤杀伤。对于PD-1和CTLA-4的免疫治疗应用,现有技术中也报道了共表达靶向肿瘤相关抗原CAR T和抑制性CAR T(胞外抗原为正常细胞抗原,胞内段为PD-1或者CTLA-4的抑制信号域),可防止CAR T脱靶现象的发生(脱靶现象:CAR T细胞识别杀伤正常细胞)。In the prior art, the application of the immunosuppressive-associated antigens PD-1 and CTLA-4 to tumor immunotherapy is usually to construct a PD-1 antibody (Pembrolizumab, Nivolumab) or a CTLA-4 antibody (Ipilimumab), or use PD- 1 or an inhibitor of CTLA-4 or a signaling pathway blocker. However, PD-1 antibodies and CTLA-4 antibodies have certain limitations, that is, antibodies need to penetrate into tumor tissues to exert therapeutic effects, so the clinical application of antibodies is uncertain. It has been reported that when CAR-GD2T cells are combined with PD-1 antibody for immunotherapy, they can resist the immunosuppressive effect of the tumor microenvironment and enhance the tumor killing of CAR T cells. For immunotherapeutic applications of PD-1 and CTLA-4, co-expression of targeting tumor-associated antigens CAR T and inhibitory CAR T has also been reported in the prior art (the extracellular antigen is a normal cell antigen and the intracellular domain is PD-1). Or the inhibitory signal domain of CTLA-4) prevents the occurrence of CAR T off-target (off-target phenomenon: CAR T cells recognize and kill normal cells).
另外,现有技术中还报道了共表达双抗体PD-1和CD19/Mesothelin/PSCA的CAR T,这虽然扩大了靶点范围(表达PD-1或CD19/Mesothelin/PSCA或同时表达两者的肿瘤细胞),但是忽略了盲目增加细胞杀伤作用所引起的副作用:细胞因子风暴、脱靶现象(正常细胞也表达两个抗原或之一,导致CAR T杀伤正常细胞,引起正常细胞缺失的机体功能缺陷)。In addition, CAR T co-expressing the diabody PD-1 and CD19/Mesothelin/PSCA has also been reported in the prior art, although this expands the target range (expressing PD-1 or CD19/Mesothelin/PSCA or both) Tumor cells), but neglecting the side effects caused by blindly increasing cell killing: cytokine storm, off-target phenomenon (normal cells also express two antigens or one, causing CAR T to kill normal cells, causing functional defects in normal cell loss) ).
发明内容Summary of the invention
本发明的目的在于提供一种嵌合的免疫抑制检查点受体分子,编码其的核酸,含有该核酸的构建体、表达载体和转化的细胞,以及它们的制药用途。本发明的表达所述嵌合的免疫抑制检查点受体分子的转化的细胞可营造一种肿瘤免疫抑制微环境,具有增强的肿瘤杀伤作用;并且,该转化的细胞有自调节功能,可缓解过度的非特异免疫效应和细胞因子风暴等副作用。 It is an object of the present invention to provide a chimeric immunosuppressive checkpoint receptor molecule, a nucleic acid encoding the same, a construct comprising the nucleic acid, an expression vector and transformed cells, and their pharmaceutical use. The transformed cell expressing the chimeric immunosuppressive checkpoint receptor molecule of the present invention can create a tumor immunosuppressive microenvironment with enhanced tumor killing effect; and the transformed cell has self-regulating function and can be alleviated Excessive side effects such as non-specific immune effects and cytokine storms.
本发明通过以下技术方案实现上述目的:The present invention achieves the above objects by the following technical solutions:
第一方面,本发明提供了一种嵌合的免疫抑制检查点受体分子,其包含胞外结构域、跨膜结构域和胞内结构域,其中,所述胞外结构域和任选地跨膜结构域为免疫抑制检查点受体分子的相应结构域或基于该结构域进行的基因改造,所述胞内结构域为一个或多个免疫激活信号结构域。In a first aspect, the invention provides a chimeric immunosuppressive checkpoint receptor molecule comprising an extracellular domain, a transmembrane domain and an intracellular domain, wherein the extracellular domain and optionally The transmembrane domain is the corresponding domain of an immunosuppressive checkpoint receptor molecule or is genetically engineered based on the domain, the intracellular domain being one or more immune activation signal domains.
作为优选,所述免疫抑制检查点受体分子选自PD-1、CTLA-4、LAG3、TIM3、A2AR、B7H3、B7H4、BTLA、IDO、KIR、CD160、2B4(CD244)和VISTA(C10orf54);Preferably, the immunosuppressive checkpoint receptor molecule is selected from the group consisting of PD-1, CTLA-4, LAG3, TIM3, A2AR, B7H3, B7H4, BTLA, IDO, KIR, CD160, 2B4 (CD244) and VISTA (C10orf54);
进一步优选地,所述免疫检查点受体分子为PD-1、CTLA-4、LAG3或TIM3。Further preferably, the immune checkpoint receptor molecule is PD-1, CTLA-4, LAG3 or TIM3.
优选地,所述胞内结构域包含免疫共刺激信号组合和任选的信号肽;Preferably, the intracellular domain comprises an immunostimulatory signal combination and an optional signal peptide;
进一步优选地,所述免疫共刺激信号组合包含TLR1和/或TLR2信号结构域;Further preferably, said immune costimulatory signal combination comprises a TLR1 and/or TLR2 signal domain;
进一步优选地,所述胞内结构域为TLR1和/或TLR2与41BB和/或CD28信号结构域的组合。Further preferably, the intracellular domain is a combination of TLR1 and/or TLR2 and a 41BB and/or CD28 signaling domain.
进一步优选地,任选的信号肽为免疫抑制检查点受体分子相对应的信号肽。Further preferably, the optional signal peptide is a signal peptide corresponding to an immunosuppressive checkpoint receptor molecule.
进一步优选地,所述胞内结构域还包括CD3ζ胞内结构域;Further preferably, the intracellular domain further comprises a CD3 sputum intracellular domain;
在具体的优选实施方案中,所述TLR1和/或TLR2信号结构域和任选地41BB和/或CD28信号结构域配置在CD3ζ胞内结构域的N末端侧。In a specific preferred embodiment, the TLR1 and/or TLR2 signal domain and optionally the 41BB and/or CD28 signal domain are arranged on the N-terminal side of the CD3 cell intracellular domain.
第二方面,本发明提供了编码如第一方面所述的嵌合的免疫抑制检查点受体分子的核酸。In a second aspect, the invention provides a nucleic acid encoding a chimeric immunosuppressive checkpoint receptor molecule of the first aspect.
第三方面,本发明提供了一种核酸构建体,其包含如第二方面所述的核酸,以及与之可操作连接、可指导所述嵌合的免疫抑制检查点受体分子在宿主细胞中表达的一个或多个控制序列。In a third aspect, the present invention provides a nucleic acid construct comprising the nucleic acid of the second aspect, and an immunosuppressive checkpoint receptor molecule operably linked thereto for directing said chimerism in a host cell One or more control sequences expressed.
第四方面,本发明提供了一种表达载体,其包含如第三方面所述的核酸构建体;优选地,所述表达载体为慢病毒表达载体。 In a fourth aspect, the present invention provides an expression vector comprising the nucleic acid construct of the third aspect; preferably, the expression vector is a lentiviral expression vector.
第五方面,本发明提供了一种转化的细胞,其包含如第二方面所述的核酸,或者其中转化了如第三方面所述的核酸构建体或如第四方面所述的表达载体;In a fifth aspect, the present invention provides a transformed cell comprising the nucleic acid of the second aspect, or wherein the nucleic acid construct of the third aspect or the expression vector of the fourth aspect is transformed;
优选地,所述转化的细胞为免疫细胞,进一步优选为T细胞、B细胞或NK细胞。Preferably, the transformed cell is an immune cell, further preferably a T cell, a B cell or an NK cell.
在具体的实施方案中,所述转化的细胞为宙斯盾细胞毒性T淋巴细胞(S-CTL,Zeushield Cytotoxic T Lymphocyte),其为过表达免疫抑制向激活转型的检查点分子的淋巴细胞,特别是T淋巴细胞。S-CTL细胞包括表达嵌合的PD-1受体分子的SP-CTL细胞、表达嵌合的CTLA-4受体分子的SC-CTL细胞、表达嵌合的TIM3受体分子的ST-CTL细胞和表达嵌合的LAG3受体分子的SL-CTL细胞等。S-CTL细胞通过在人体T细胞中转染嵌合的免疫抑制-激动转型的检查点分子基因,获得可抵抗免疫抑制的淋巴细胞。S-CTL淋巴细胞具有以下特点:In a specific embodiment, the transformed cell is a Zeus sulphate cytotoxic T lymphocyte (S-CTL), which is a lymphocyte that overexpresses a checkpoint molecule that undergoes immunosuppression to activation, particularly T lymphocytes. S-CTL cells include SP-CTL cells expressing a chimeric PD-1 receptor molecule, SC-CTL cells expressing a chimeric CTLA-4 receptor molecule, and ST-CTL cells expressing a chimeric TIM3 receptor molecule. And SL-CTL cells expressing a chimeric LAG3 receptor molecule and the like. S-CTL cells obtain lymphocytes resistant to immunosuppression by transfecting a chimeric immunosuppressive-agonistic transition checkpoint molecule gene into human T cells. S-CTL lymphocytes have the following characteristics:
1)对表达免疫抑制检查点分子的配体(如PD-L1、B7.1、B7.2、Galectin-9、MHC II等)的肿瘤细胞或肿瘤相关免疫抑制细胞具有强效细胞杀伤作用;1) potent cell killing effect on tumor cells or tumor-associated immunosuppressive cells expressing ligands of immunosuppressive checkpoint molecules (such as PD-L1, B7.1, B7.2, Galectin-9, MHC II, etc.);
2)S-CTL包含天然的免疫抑制检查点受体分子(如PD-1、CTLA-4、TIM3、LAG3等)表达在免疫效应细胞特别是T细胞、NK细胞等表面,与肿瘤细胞或肿瘤相关免疫抑制细胞的配体结合后,可激活S-CTL载体下游的免疫激活信号,促进免疫细胞自身的激活、增殖、识别、杀伤等功能的发挥;2) S-CTL contains natural immunosuppressive checkpoint receptor molecules (such as PD-1, CTLA-4, TIM3, LAG3, etc.) expressed on the surface of immune effector cells, especially T cells, NK cells, etc., and tumor cells or tumors. After binding of the ligand of the relevant immunosuppressive cell, the immune activation signal downstream of the S-CTL vector can be activated to promote the activation, proliferation, recognition, killing and other functions of the immune cell itself;
3)S-CTL结构:胞外段和跨膜区域为完全天然的免疫抑制检查点分子结构,用于特异性识别肿瘤细胞表面配体及识别信号在S-CTL细胞内的有效传递;胞内区域为一个或多个免疫激活信号域,其中本发明发现含TLR1和/或TLR2信号激活域的胞内结构域,可进一步促进肿瘤杀伤效应;3) S-CTL structure: extracellular segment and transmembrane region are completely natural immunosuppressive checkpoint molecular structures for specific recognition of tumor cell surface ligands and recognition signals for efficient delivery in S-CTL cells; intracellular The region is one or more immune activation signal domains, wherein the present invention finds an intracellular domain comprising a TLR1 and/or TLR2 signaling activation domain, which further promotes tumor killing effects;
4)自调节功能,由于正常淋巴细胞表达天然的免疫抑制检查点分子受体,转化的细胞表达嵌合的免疫抑制-激动转型的检查点分子,而天然的免疫抑制检查点分子受体为传导抑制信号,嵌合的免疫抑制检查点分子受体为传导激活信号,由此可通过免疫 效应细胞自身进行反馈表达调控,减缓S-CTL细胞过度激活引起的副作用,如脱靶杀伤、细胞因子风暴;4) Self-regulating function, since normal lymphocytes express natural immunosuppressive checkpoint molecular receptors, transformed cells express chimeric immunosuppressive-agonistic transition checkpoint molecules, while natural immunosuppressive checkpoint molecular receptors are conductive Inhibition of the signal, the chimeric immunosuppressive checkpoint molecule receptor is a conduction activation signal, thereby allowing immunity The effector cells themselves perform feedback expression regulation and slow down the side effects caused by excessive activation of S-CTL cells, such as off-target killing and cytokine storm;
第六方面,本发明提供了制备如第五方面所述的转化的细胞的方法,其包括:向细胞中引入如第二方面所述的核酸,或者转化如第三方面所述的核酸构建体或如第四方面所述的表达载体的步骤。In a sixth aspect, the invention provides a method of producing a transformed cell of the fifth aspect, comprising: introducing a nucleic acid of the second aspect into a cell, or transforming the nucleic acid construct of the third aspect Or the step of expressing the vector as described in the fourth aspect.
第七方面,本发明提供了如第一方面所述的嵌合的免疫抑制检查点受体分子、如第二方面所述的核酸、如第三方面所述的核酸构建体、如第四方面所述的表达载体或如第五方面所述的转化的细胞在制备与免疫抑制检查点受体分子对应的配体、优选与PD-L1、B7.1、B7.2、Galectin-9、MHC II的表达相关的疾病的药物中的用途;In a seventh aspect, the invention provides a chimeric immunosuppressive checkpoint receptor molecule according to the first aspect, a nucleic acid according to the second aspect, a nucleic acid construct according to the third aspect, the fourth aspect The expression vector or the transformed cell of the fifth aspect, in the preparation of a ligand corresponding to an immunosuppressive checkpoint receptor molecule, preferably with PD-L1, B7.1, B7.2, Galectin-9, MHC Use of a drug for expression of a disease associated with II;
优选地,所述疾病为表达免疫抑制检查点受体分子对应的配体、优选表达PD-L1、B7.1、B7.2、Galectin-9、MHC II的肿瘤;Preferably, the disease is a tumor corresponding to a ligand expressing an immunosuppressive checkpoint receptor molecule, preferably a tumor expressing PD-L1, B7.1, B7.2, Galectin-9, MHC II;
进一步优选地,所述表达免疫抑制检查点受体分子对应的配体、优选表达PD-L1、B7.1、B7.2、Galectin-9、MHC II的肿瘤选自肺癌、白血病、乳腺癌、前列腺癌、胰腺癌、肝癌、黑色素瘤和非黑色素瘤皮肤癌。Further preferably, the tumor corresponding to the immunosuppressive checkpoint receptor molecule, preferably expressing PD-L1, B7.1, B7.2, Galectin-9, MHC II, is selected from the group consisting of lung cancer, leukemia, breast cancer, Prostate cancer, pancreatic cancer, liver cancer, melanoma, and non-melanoma skin cancer.
术语定义Definition of Terms
在本发明的上下文中,“免疫检查点”(Immune Checkpoint)是免疫系统中可上调或下调免疫刺激信号的分子,分为免疫刺激检查点分子(Stimulatory Checkpoint)和免疫抑制检查点分子(Inhibitory Checkpoint)。In the context of the present invention, an "Immune Checkpoint" is a molecule that up-regulates or down-regulates an immunostimulatory signal in the immune system, and is classified into a Stimulatory Checkpoint and an Inhibitory Checkpoint molecule. ).
“免疫抑制检查点”是免疫系统中可下调免疫刺激信号的分子,例如:PD-1、CTLA-4、TIM3、LAG3、A2AR、B7-H3、B7-H4、BTLA、IDO、KIR、CD160、2B4(CD244)、VISTA(C10orf54)、TIGIT、LAIR1等。An "immunosuppressive checkpoint" is a molecule that down-regulates an immune stimulus signal in the immune system, such as: PD-1, CTLA-4, TIM3, LAG3, A2AR, B7-H3, B7-H4, BTLA, IDO, KIR, CD160, 2B4 (CD244), VISTA (C10orf54), TIGIT, LAIR1, etc.
“免疫激活信号”:淋巴细胞激活过程中,T细胞需要获取两个信号以充分激活;第一信号是抗原特异性的,即通过T细胞受体与抗原递呈细胞APC表面MHC分子结 合产生的;第二信号,即共刺激信号,是抗原非特异性的,通过T细胞和APC细胞表面的共刺激信号分子结合产生的。"Immune activation signal": During lymphocyte activation, T cells need to acquire two signals for full activation; the first signal is antigen-specific, that is, through the T cell receptor and the antigen-presenting cell APC surface MHC molecule junction The second signal, the costimulatory signal, is antigen-unspecific and is produced by the binding of T cells and costimulatory signaling molecules on the surface of APC cells.
本发明的嵌合的免疫抑制检查点受体分子的胞内结构域可以包括共刺激信号分子CD28、OX40、ICOS(CD278)、4-1BB(CD137)、CD40、CD27、CD134、CDS、ICAM-1、LFA-1(CD11a/CD18)、CD2、BTLA等的胞内段,或者它们的任意组合。The intracellular domain of the chimeric immunosuppressive checkpoint receptor molecule of the invention may comprise costimulatory signaling molecules CD28, OX40, ICOS (CD278), 4-1BB (CD137), CD40, CD27, CD134, CDS, ICAM- 1. Intracellular segments of LFA-1 (CD11a/CD18), CD2, BTLA, etc., or any combination thereof.
有益效果Beneficial effect
本发明所述转化的细胞是一种新型的肿瘤杀伤细胞,其特异性识别杀伤肿瘤免疫抑制微环境中表达免疫抑制检查点分子配体的细胞,具有低脱靶率;并且,其胞内结构域的独特设计可进一步促进其肿瘤杀伤效应,同时可以有效消除肿瘤免疫抑制作用;而且,本发明所转化的免疫效应细胞(如T细胞等)处于肿瘤微环境中会诱导表达一定量的天然免疫抑制检查点受体分子,其与肿瘤细胞、肿瘤相关免疫抑制细胞或免疫自身表达的抗原结合产生一定的反馈抑制调节信号,对转化的细胞产生的免疫效应有一定的调节作用,即不会导致所转化的细胞过渡激活,引起炎症等不适症状。此外,本发明所转化的激活载体进行了免疫激活优化,包含固有免疫激活信号TLR1/TLR2胞内段,且组合4-1BB、CD28等免疫激活分子胞内段构建共刺激信号,最后联合CD3ζ链构建完整的免疫信号传导,具有极强的免疫激活效果,S-CTL细胞具有极强的杀伤肿瘤细胞、肿瘤相关免疫抑制细胞的功能,如SC-CTL细胞可特异性识别杀伤肿瘤微环境中的CD80+/CD86+肿瘤细胞和肿瘤相关免疫抑制细胞,协同促进抗肿瘤免疫效应。The transformed cell of the present invention is a novel tumor killer cell which specifically recognizes cells expressing an immunosuppressive checkpoint molecular ligand in a tumor suppressing immunosuppressive microenvironment, has a low off-target rate; and, its intracellular domain The unique design can further promote its tumor killing effect, and can effectively eliminate the tumor immunosuppressive effect; Moreover, the immune effector cells (such as T cells, etc.) transformed by the present invention are induced to express a certain amount of natural immunosuppression in the tumor microenvironment. Checkpoint receptor molecules, which bind to tumor cells, tumor-associated immunosuppressive cells, or immune-expressed antigens to produce a certain feedback inhibition regulatory signal, which has a certain regulatory effect on the immune effects produced by transformed cells, ie, does not cause The transformed cells are transiently activated, causing discomfort such as inflammation. In addition, the activated vector transformed by the present invention is optimized for immune activation, and comprises an intrinsic immune activation signal TLR1/TLR2 intracellular segment, and a combination of 4-1BB, CD28 and other immune activation molecules to construct a co-stimulatory signal, and finally a CD3 ζ chain The construction of complete immune signaling has a strong immune activation effect. S-CTL cells have strong functions of killing tumor cells and tumor-associated immunosuppressive cells. For example, SC-CTL cells can specifically recognize the microenvironment in killing tumors. CD80+/CD86+ tumor cells and tumor-associated immunosuppressive cells synergistically promote anti-tumor immune effects.
鉴于上述优势,本发明的表达所述嵌合受体分子的转化的细胞在抗肿瘤治疗中具有良好的临床应用前景。In view of the above advantages, the transformed cells expressing the chimeric receptor molecule of the present invention have a good clinical application prospect in antitumor therapy.
附图说明DRAWINGS
图1显示实施例1中的合成序列图谱。 Figure 1 shows the synthetic sequence map in Example 1.
图2显示实施例1中构建的pWPXLd-S(PD1)-CTL-2A-EGFP质粒、pWPXLd-S(CTLA-4)-CTL-2A-EGFP质粒、pWPXLd-S(TIM3)-CTL-2A-EGFP质粒和pWPXLd-S(LAG3)-CTL-2A-EGFP质粒图谱。Figure 2 shows the pWPXLd-S(PD1)-CTL-2A-EGFP plasmid constructed in Example 1, pWPXLd-S(CTLA-4)-CTL-2A-EGFP plasmid, pWPXLd-S(TIM3)-CTL-2A- EGFP plasmid and pWPXLd-S (LAG3)-CTL-2A-EGFP plasmid map.
图3显示流式细胞仪检测SP-CTL细胞的病毒感染阳性率;空白组:不使用CD3流式抗体染色的非转染T细胞。Figure 3 shows the positive rate of viral infection of SP-CTL cells by flow cytometry; blank group: non-transfected T cells stained without CD3 flow cytometry.
图4显示流式细胞仪检测SC-CTL细胞的病毒感染阳性率;正常T细胞组:非转染T细胞。Figure 4 shows the positive rate of viral infection of SC-CTL cells by flow cytometry; normal T cell group: non-transfected T cells.
图5显示SP-CTL细胞对人T-ALL细胞系JURKAT-GL的杀伤作用。Figure 5 shows the killing effect of SP-CTL cells on the human T-ALL cell line JURKAT-GL.
图6显示SP-CTL细胞对人非小细胞性肺癌H460-GL的杀伤作用。Figure 6 shows the killing effect of SP-CTL cells on human non-small cell lung cancer H460-GL.
图7显示SC-CTL细胞对人B-ALL细胞系NALM6-GL杀伤作用。Figure 7 shows the killing effect of SC-CTL cells on the human B-ALL cell line NALM6-GL.
图8显示SP-CTL细胞对人非小细胞性肺癌H460-GL的体内杀伤作用。Figure 8 shows the in vivo killing effect of SP-CTL cells on human non-small cell lung cancer H460-GL.
图9显示SP-CTL细胞对不表达PD-L1的A549细胞的体外杀伤作用。Figure 9 shows the in vitro killing effect of SP-CTL cells on A549 cells that do not express PD-L1.
图10显示SP-CTL细胞对不表达PD-L1的K562细胞的体外杀伤作用。Figure 10 shows the in vitro killing effect of SP-CTL cells on K562 cells that do not express PD-L1.
图11显示流式细胞术检测PD-L1在肺癌细胞系H1299、H460、H23和A549以及前列腺癌细胞系PC3中的表达情况的结果图。Figure 11 is a graph showing the results of flow cytometry detection of PD-L1 expression in lung cancer cell lines H1299, H460, H23 and A549 and prostate cancer cell line PC3.
图12显示SP-CTL细胞对表达PD-L1的肺癌细胞系H1299、H460、H23和A549以及前列腺癌细胞系PC3的体外杀伤作用。Figure 12 shows the in vitro killing effect of SP-CTL cells on PD-L1 expressing lung cancer cell lines H1299, H460, H23 and A549 and prostate cancer cell line PC3.
图13显示SP-CTL细胞体内识别、杀伤原代肺癌细胞的结果图;其中图13-A显示在不同时间点,各实验组肿瘤组织块的平均体积结果,图13-B显示每个实验组三个重复的肿瘤组织块的尺寸比对图片。Figure 13 is a graph showing the results of in vivo recognition and killing of primary lung cancer cells by SP-CTL cells; wherein Fig. 13-A shows the average volume results of tumor tissue blocks in each experimental group at different time points, and Fig. 13-B shows each experimental group. The size of the three replicated tumor tissue blocks is aligned with the picture.
图14显示SP-CTL细胞体内识别、杀伤原代胃癌细胞的结果图;其中图14-A显示每个实验组三个重复的肿瘤组织块的尺寸比对图片,图14-B显示各实验组的肿瘤组织块的平均重量结果。 Figure 14 is a graph showing the results of in vivo recognition and killing of primary gastric cancer cells by SP-CTL cells; wherein Figure 14-A shows the size comparison of three replicate tumor tissue blocks in each experimental group, and Figure 14-B shows the experimental groups. The average weight result of the tumor tissue mass.
图15显示SP-CTL细胞体内识别、杀伤原代骨肉瘤细胞的结果图;其中图15-A显示每个实验组六个重复的肿瘤组织块的尺寸比对图片,图15-B显示各实验组的肿瘤组织块的平均重量结果。Figure 15 is a graph showing the results of in vivo recognition and killing of primary osteosarcoma cells by SP-CTL cells; wherein Figure 15-A shows a size comparison of six replicate tumor tissue blocks in each experimental group, and Figure 15-B shows each experiment. The average weight result of the group of tumor tissue blocks.
图16显示SC-CTL细胞体外特异性识别、杀伤非霍金森淋巴瘤RL细胞的结果图。Figure 16 is a graph showing the results of in vitro specific recognition and killing of non-Hawsonson lymphoma RL cells by SC-CTL cells.
图17显示SC-CTL细胞体内特异性识别、杀伤非霍金森淋巴瘤RL细胞的结果图;其中图17-A显示每个实验组两个重复的肿瘤组织块的尺寸比对图片,图17-B显示每个实验组的肿瘤组织块的平均重量结果。Figure 17 is a graph showing the results of in vivo recognition and killing of non-Hawking's lymphoma RL cells by SC-CTL cells; Figure 17-A shows a comparison of the size of two replicate tumor tissue blocks in each experimental group, Figure 17- B shows the average weight result of the tumor tissue mass of each experimental group.
图18显示mSC-CTL细胞体内特异性识别、杀伤原代骨肉瘤细胞的结果图;其中图18-A显示每个实验组的肿瘤组织块的尺寸比对图片,图18-B显示各实验组的肿瘤组织块的平均重量结果。Figure 18 is a graph showing the results of in vivo recognition and killing of primary osteosarcoma cells by mSC-CTL cells; wherein Figure 18-A shows the size comparison of the tumor tissue blocks of each experimental group, and Figure 18-B shows the experimental groups. The average weight result of the tumor tissue mass.
图19显示SP-CTL、mSC-CTL单独或联合应用在体内特异性识别、杀伤原代肺癌细胞的结果图;其中图19-A显示各实验组肿瘤组织块的平均体积结果,图19-B显示各实验组肿瘤组织块的平均重量结果。Figure 19 is a graph showing the results of SP-CTL, mSC-CTL alone or in combination to specifically recognize and kill primary lung cancer cells in vivo; Figure 19-A shows the average volume results of tumor tissue blocks in each experimental group, Figure 19-B The average weight results of the tumor tissue blocks of each experimental group are shown.
图20显示SP-CTL、mSC-CTL单独或联合应用在体内对肿瘤组织中的CD80+细胞(图20-A)、CD86+细胞(图20-B)及CD11b+Ly6G+细胞(MDSC)(图20-C)的影响。Figure 20 shows that SP-CTL, mSC-CTL alone or in combination with CD80+ cells (Fig. 20-A), CD86+ cells (Fig. 20-B) and CD11b+Ly6G+ cells (MDSC) in tumor tissues in vivo (Fig. 20- C) The impact.
图21显示B急性淋巴细胞白血病细胞系Nalm6、RL、Raji高表达Lag3配体MHC II。Figure 21 shows that the B acute lymphoblastic leukemia cell line Nalm6, RL, Raji highly expresses the Lag3 ligand MHC II.
图22显示SL-CTL细胞可特异性识别和杀伤高表达MHC II配体的B-ALL细胞系Nalm6细胞。Figure 22 shows that SL-CTL cells specifically recognize and kill the B-ALL cell line Nalm6 cells that highly express MHC II ligand.
图23显示与肿瘤细胞共孵育的SL-CTL细胞可显著上调炎症因子IL-2(图23A)、IFN-γ(图23A)的蛋白表达和分泌。Figure 23 shows that SL-CTL cells co-incubated with tumor cells significantly up-regulated protein expression and secretion of inflammatory factors IL-2 (Figure 23A), IFN-γ (Figure 23A).
图24显示A549细胞在体外不表达Galectin9(Tim3配体)表面蛋白,而体内微 环境可诱导肺癌细胞系A549特异性高表达Galectin9表面蛋白。Figure 24 shows that A549 cells do not express Galectin9 (Tim3 ligand) surface protein in vitro, but in vivo micro The environment-induced lung cancer cell line A549 specifically expresses Galectin9 surface protein.
图25显示原代肺癌细胞1、2、3在病人来源的肿瘤异种移植(PDX,Patient Derived Xenograft)模型中特异性高表达Galectin9表面蛋白。Figure 25 shows that primary lung cancer cells 1, 2, and 3 specifically express Galectin9 surface protein in a patient-derived tumor xenograft (PDX) model.
图26显示ST-CTL可在PDX模型中特异性识别杀伤高表达Galectin9表面蛋白的原代肺癌细胞。Figure 26 shows that ST-CTL specifically recognizes primary lung cancer cells that kill high expression of Galectin9 surface protein in the PDX model.
具体实施方式detailed description
为便于理解本发明,本发明列举实施例如下。本领域技术人员应该明了,所述实施例仅仅是帮助理解本发明,不应视为对本发明的具体限制。To facilitate an understanding of the invention, the invention is set forth below. It should be understood by those skilled in the art that the present invention is not to be construed as limited.
实施例1、S-CTL细胞的制备Example 1. Preparation of S-CTL cells
质粒构建Plasmid construction
1)通过基因合成,得到“免疫抑制检查点分子(信号肽+胞外段+跨膜区)-免疫刺激信号1胞内段-免疫刺激信号2(一般为TLR1或者TLR2)胞内段-CD3ζ”的DNA序列,DNA序列的首尾分别包含PmeI和Spe1限制性酶切位点。合成序列图谱如图1所示。1) Through gene synthesis, "immunosuppressive checkpoint molecule (signal peptide + extracellular segment + transmembrane region) - immunostimulatory signal 1 intracellular segment - immunostimulatory signal 2 (generally TLR1 or TLR2) intracellular segment - CD3 得到The DNA sequence, the first and the end of the DNA sequence contain the PmeI and Spe1 restriction sites, respectively. The synthetic sequence map is shown in Figure 1.
本发明实施例中所合成的基因序列请见SEQ NO.1(为制备靶向PD-1的S-CTL,即SP-CTL,而合成的DNA序列,结构为PD-1信号肽+胞外段+跨膜区-CD28-TLR1/2-CD3ζ)和SEQ NO.2(为制备靶向CTLA-4的S-CTL,即SC-CTL,而合成的DNA序列,结构为CTLA-4信号肽+胞外段+跨膜区-CD28胞内段-TLR1/2胞内段-CD3ζ)和SEQ NO.3(为制备靶向TIM3的S-CTL,即ST-CTL,而合成的DNA序列,结构为TIM3信号肽+胞外段+跨膜区-4-1BB胞内段-TLR2胞内段-CD3ζ)和SEQ NO.4(为制备靶向LAG3的S-CTL,即SL-CTL,而合成的DNA序列,结构为LAG3信号肽+胞外段+跨膜区-4-1BB胞内段-TLR2胞内段-CD3ζ)。For the gene sequence synthesized in the examples of the present invention, see SEQ NO. 1 (for the preparation of S-CTL targeting PD-1, ie, SP-CTL, the synthesized DNA sequence, the structure is PD-1 signal peptide + extracellular Segment + transmembrane region - CD28-TLR1/2-CD3ζ) and SEQ NO. 2 (for the preparation of S-CTL targeting CTLA-4, ie, SC-CTL, the synthesized DNA sequence, the structure is CTLA-4 signal peptide + extracellular segment + transmembrane region - CD28 intracellular segment - TLR1/2 intracellular segment - CD3 ζ) and SEQ NO. 3 (for the preparation of S-CTL targeting TIM3, ie, ST-CTL, the synthesized DNA sequence, The structure is TIM3 signal peptide + extracellular domain + transmembrane region - 4-1BB intracellular segment - TLR2 intracellular segment - CD3 ζ) and SEQ NO. 4 (for the preparation of S-CTL targeting LAG3, ie SL-CTL, The synthesized DNA sequence has the structure of LAG3 signal peptide + extracellular segment + transmembrane region - 4-1BB intracellular segment - TLR2 intracellular segment - CD3 ζ).
S-CTL(Zeushield Cytotoxic T Lymphocyte)细胞,为基因修饰过表达S-CTL嵌合 受体以增强免疫杀伤能力的T淋巴细胞,同时不仅局限于T细胞,过表达S-CTL嵌合受体的免疫效应细胞亦具有增强的免疫杀伤功能。S-CLT嵌合受体,即嵌合的免疫抑制检查点分子,胞外段和跨膜段为天然的人免疫抑制检查点受体分子如PD-1、CTLA-4、TIM3、LAG3、A2AR、B7-H3、B7-H4、BTLA、IDO、KIR、CD160、2B4(CD244)、VISTA(C10orf54)、TIGIT、LAIR1等的相应区段,而胞内段为免疫共刺激信号分子包含TLR1/TLR2与TLR3-10、4-1BB、CD28、ICOS、CD27、CD2、OX40、CD30、CD40、LFA-1、CD7、CD2S、LIGHT、NKG2C、B7-H3、CD83等胞内段的组合,及其与CD3zeta链连接组成;可特异性识别和杀伤表达免疫抑制检查点配体的肿瘤细胞和肿瘤相关的免疫抑制细胞。S-CTL (Zeushield Cytotoxic T Lymphocyte) cells, which are genetically modified to overexpress S-CTL chimera Receptors are T lymphocytes that enhance immune killing ability, and are not limited to T cells, and immune effector cells overexpressing S-CTL chimeric receptors also have enhanced immunological killing function. S-CLT chimeric receptors, ie chimeric immunosuppressive checkpoint molecules, extracellular and transmembrane segments are natural human immunosuppressive checkpoint receptor molecules such as PD-1, CTLA-4, TIM3, LAG3, A2AR , B7-H3, B7-H4, BTLA, IDO, KIR, CD160, 2B4 (CD244), VISTA (C10orf54), TIGIT, LAIR1, etc., and the intracellular segment is an immunostimulatory signaling molecule comprising TLR1/TLR2 Combination with intracellular segments such as TLR3-10, 4-1BB, CD28, ICOS, CD27, CD2, OX40, CD30, CD40, LFA-1, CD7, CD2S, LIGHT, NKG2C, B7-H3, CD83, etc. The CD3zeta chain is composed of a combination; it can specifically recognize and kill tumor cells and tumor-associated immunosuppressive cells expressing immunosuppressive checkpoint ligands.
SP-CTL,表达嵌合的PD-1受体的S-CTL细胞,特异性识别杀伤表达PD-L1的肿瘤细胞和肿瘤相关基质细胞或免疫抑制细胞。SP-CTL, an S-CTL cell expressing a chimeric PD-1 receptor, specifically recognizes tumor cells and tumor-associated stromal cells or immunosuppressive cells that secrete PD-L1.
SC-CTL,表达嵌合的CTLA-4受体的S-CTL细胞,特异性识别杀伤表达B7.1和/或B7.2的肿瘤细胞和肿瘤相关基质细胞或免疫抑制细胞。SC-CTL, a S-CTL cell expressing a chimeric CTLA-4 receptor, specifically recognizes tumor cells and tumor-associated stromal cells or immunosuppressive cells that kill B7.1 and/or B7.2.
ST-CTL,表达嵌合的TIM3受体的S-CTL细胞,特异性识别杀伤表达Galectin-9的肿瘤细胞和肿瘤相关基质细胞或免疫抑制细胞。ST-CTL, an S-CTL cell expressing a chimeric TIM3 receptor, specifically recognizes tumor cells and tumor-associated stromal cells or immunosuppressive cells that secrete Galectin-9.
SL-CTL,表达嵌合的LAG3受体的S-CTL细胞,特异性识别杀伤表达MHC II的肿瘤细胞和肿瘤相关基质细胞或免疫抑制细胞。SL-CTL, a S-CTL cell expressing a chimeric LAG3 receptor, specifically recognizes tumor cells that secrete MHC II and tumor-associated stromal cells or immunosuppressive cells.
2)使用Thermo限制性内切酶Pme1和Spe1,分别通过双酶切获得合成的S-CTL DNA片段(SEQ NO.1、SEQ NO.2、SEQ NO.3、SEQ NO.4)和pWPXLd-EGFP载体的线性化DNA(含粘性末端)。2) Using the Thermo restriction enzymes Pme1 and Spe1, the synthetic S-CTL DNA fragments (SEQ NO. 1, SEQ NO. 2, SEQ NO. 3, SEQ NO. 4) and pWPXLd- were respectively obtained by double digestion. Linearized DNA (containing sticky ends) of the EGFP vector.
3)通过琼脂凝胶电泳回收,获得带粘性末端的线性化合成的S-CTL(SEQ NO.1、SEQ NO.2、SEQ NO.3、SEQ NO.4)和pWPXLd-EGFP载体DNA片段。3) Recovery by agarose gel electrophoresis, linearized synthetic S-CTL (SEQ NO. 1, SEQ NO. 2, SEQ NO. 3, SEQ NO. 4) and pWPXLd-EGFP vector DNA fragment with sticky ends were obtained.
4)通过T4 DNA连接酶(来自Invitrogent公司),分别连接线性化的SEQ NO.1 和pWPXLd-2A-EGFP、线性化的SEQ NO.2和pWPXLd-2A-EGFP,获得pWPXLd-S(PD1/CTLA-4/TIM3/LAG3)-CTL-2A-EGFP质粒,其质粒图谱参见附图2。4) Linked linearized SEQ NO.1 by T4 DNA ligase (from Invitrogent) pWPXLd-S(PD1/CTLA-4/TIM3/LAG3)-CTL-2A-EGFP plasmid was obtained with pWPXLd-2A-EGFP, linearized SEQ NO.2 and pWPXLd-2A-EGFP, and the plasmid map is shown in the attached drawing. 2.
S-CTL慢病毒包装S-CTL Lentiviral Packaging
1)培养293T细胞至密度达80-90%,将培养基更换为:DMEM高糖培养基+1%FBS+1%双抗;1) Incubate 293T cells to a density of 80-90%, and replace the medium with: DMEM high glucose medium + 1% FBS + 1% double antibody;
2)2-6小时后,用PEI分别将pWPXLd-S(PD1/CTLA-4/TIM3/LAG3)-CTL-2A-EGFP质粒与慢病毒辅助质粒(pMD2.G、psPAX2)共同转染入293T细胞,以空载体pWPXLd-EGFP与辅助质粒共转染作为对照组;2) After 2-6 hours, the pWPXLd-S (PD1/CTLA-4/TIM3/LAG3)-CTL-2A-EGFP plasmid was transfected into the 293T together with the lentiviral helper plasmid (pMD2.G, psPAX2) with PEI. The cells were co-transfected with the empty vector pWPXLd-EGFP and the helper plasmid as a control group;
3)分别于转染后24、48和72小时,收集培养基上清,即S(PD1/CTLA-4/TIM3/LAG3)-CTL病毒上清,冻存于-80℃备用。3) The culture supernatant, S(PD1/CTLA-4/TIM3/LAG3)-CTL virus supernatant, was collected at 24, 48 and 72 hours after transfection, and stored at -80 °C until use.
T细胞激活和慢病毒感染T cell activation and lentiviral infection
1)通过Ficoll密度梯度法或红细胞裂解法分离出血液中的单核细胞;1) separating monocytes in blood by Ficoll density gradient method or red blood cell lysis method;
2)通过MACS Pan-T磁珠分选出T细胞,用T细胞培养基(AIM-V加5%FBS、青霉素100U/ml和链霉素0.1mg/ml)稀释至浓度2.5×106个/ml待用;2) T cells were sorted by MACS Pan-T magnetic beads and diluted to a concentration of 2.5×10 6 with T cell medium (AIM-V plus 5% FBS, penicillin 100 U/ml and streptomycin 0.1 mg/ml). /ml to be used;
3)通过包被CD2、CD3、CD28抗体的磁珠(美天旎)刺激T细胞,于37℃、5%CO2条件下的培养箱中培养刺激48h;3) stimulating T cells by magnetic beads coated with CD2, CD3, and CD28 antibodies, and stimulating for 48 hours in an incubator at 37 ° C under 5% CO 2 ;
4)去除T细胞中磁珠,离心去上清,重悬;4) remove the magnetic beads in the T cells, centrifuge to remove the supernatant, and resuspend;
5)加入S(PD1/CTLA-4/TIM3/LAG3)-CTL慢病毒上清和8μg/ml的polybrene和300IU/ml IL-2,病毒加入量为MOI=10,于37℃,5%CO2条件下的培养箱中培养;5) Add S (PD1/CTLA-4/TIM3/LAG3)-CTL lentiviral supernatant and 8 μg/ml of polybrene and 300 IU/ml IL-2, the amount of virus added is MOI=10, at 37 ° C, 5% CO 2 Culture in an incubator under conditions;
6)24h后,300g离心5min,去上清,用含300IU/ml IL-2的新鲜T细胞培养基重悬,于37℃、5%CO2条件下的培养箱中培养,每2-3天进行半量换液;6) After 24 h, centrifuge at 300 g for 5 min, remove the supernatant, resuspend in fresh T cell medium containing 300 IU/ml IL-2, and incubate in an incubator at 37 ° C under 5% CO 2 for 2-3 Half a day of liquid change;
7)通过流式细胞技术,检测GFP阳性T细胞S-CTL(SC-CTL或ST-CTL)比例,结果如图3、图4所示。 7) The ratio of S-CTL (SC-CTL or ST-CTL) of GFP-positive T cells was detected by flow cytometry, and the results are shown in Fig. 3 and Fig. 4.
实施例2、SP-CTL细胞对人白血病细胞系JURKAT的体外杀伤效应Example 2: In vitro killing effect of SP-CTL cells on human leukemia cell line JURKAT
1)在白血病细胞系JURKAT中转导表达荧光素酶(Luciferace),构建JURKAT-GL报告细胞系;1) Transduction of luciferase (Luciferace) in the leukemia cell line JURKAT to construct a JURKAT-GL reporter cell line;
2)分别将SC-CTL GFP阳性细胞与JURKAT-GL细胞以细胞数1∶2比例混合共培养(对照组为加GFP T细胞,空白组为不加T细胞,每组设置三个复孔);2) SC-CTL GFP-positive cells and JURKAT-GL cells were mixed and cultured in a ratio of 1:2 (the control group was GFP T cells, the blank group was no T cells, and each group was set with three duplicate wells). ;
3)18小时后,轻轻用移液器吸除一半体积的培养基上清,加入等体积的荧光素酶底物(浓度:300μg/ml),混匀;3) After 18 hours, gently pipette a half volume of the medium supernatant, add an equal volume of luciferase substrate (concentration: 300 μg / ml), and mix;
4)通过多功能酶标仪测定RLU(relative light unit),测定时间设为1秒。4) The RLU (relative light unit) was measured by a multi-function microplate reader, and the measurement time was set to 1 second.
5)杀伤比例计算公式:100%×(空白孔读数-实验孔读数)/空白孔读数,SP-CTL细胞对人白血病细胞系JURKAT的体外杀伤比例如图5所示。5) Formula for killing ratio: 100% × (blank hole reading - experimental hole reading) / blank hole reading, the in vitro killing ratio of SP-CTL cells to human leukemia cell line JURKAT is shown in FIG. 5 .
实施例3、SP-CTL细胞对非小细胞性肺癌H460的体外杀伤效应Example 3: In vitro killing effect of SP-CTL cells on non-small cell lung cancer H460
1)在非小细胞性肺癌H460中转导表达荧光素酶(Luciferace),构建H460-GL报告细胞系;1) Transduction of luciferase (Luciferace) in non-small cell lung cancer H460 to construct a H460-GL reporter cell line;
2)分别将SP-CTL GFP阳性细胞与H460-GL细胞以细胞数1∶2比例混合共培养(对照组为加GFP T细胞,空白组为不加T细胞,每组设置三个复孔);2) SP-CTL GFP-positive cells and H460-GL cells were mixed and co-cultured at a ratio of 1:2 (the control group was GFP T cells, the blank group was no T cells, and each group was set with three duplicate wells). ;
3)18小时后,轻轻用移液器吸除一半体积的培养基上清,加入等体积的荧光素酶底物(浓度:300μg/ml),混匀;3) After 18 hours, gently pipette a half volume of the medium supernatant, add an equal volume of luciferase substrate (concentration: 300 μg / ml), and mix;
4)通过多功能酶标仪测定RLU(relative light unit),,测定时间设为1秒。4) The RLU (relative light unit) was measured by a multi-function microplate reader, and the measurement time was set to 1 second.
5)杀伤比例计算公式:100%×(空白孔读数-实验孔读数)/空白孔读数(如图四),SP-CTL细胞对非小细胞性肺癌H460的体外杀伤细胞比例如图6所示。5) Formula for killing ratio: 100% × (blank hole reading - experimental hole reading) / blank hole reading (Figure 4), the ratio of SP-CTL cells to non-small cell lung cancer H460 in vitro is shown in Figure 6. .
实施例4、SC-CTL细胞对人白血病细胞系NALM6的体外杀伤效应Example 4: In vitro killing effect of SC-CTL cells on human leukemia cell line NALM6
1)在白血病细胞系NALM6中转导表达荧光素酶(Luciferace),构建NALM6-GL报告细胞系; 1) Transduction of luciferase (Luciferace) in the leukemia cell line NALM6 to construct a NALM6-GL reporter cell line;
2)分别将GFP阳性细胞数为3.5×104、1.75×104、8.6×103、4.4×103、2.2×103、1.1×103的SC-CTL病毒感染混合细胞与1×104个NALM6-GL细胞共培养(对照组为加GFP T细胞,空白组为不加T细胞,每组设置三个复孔);2) SC-CTL virus-infected mixed cells with 1×10 of GFP-positive cells were 3.5×10 4 , 1.75×10 4 , 8.6×10 3 , 4.4×10 3 , 2.2×10 3 , and 1.1×10 3 , respectively . Four NALM6-GL cells were co-cultured (the control group was GFP T cells, the blank group was no T cells, and each group was set with three duplicate wells);
3)18小时后,轻轻用移液器吸除一半体积的培养基上清,加入等体积的荧光素酶底物(浓度:300μg/ml),混匀;3) After 18 hours, gently pipette a half volume of the medium supernatant, add an equal volume of luciferase substrate (concentration: 300 μg / ml), and mix;
4)通过多功能酶标仪测定RLU(relative light unit),测定时间设为1秒。4) The RLU (relative light unit) was measured by a multi-function microplate reader, and the measurement time was set to 1 second.
5)杀伤比例计算公式:100%×(空白孔读数-实验孔读数)/空白孔读数,SC-CTL细胞对人白血病细胞系NALM6的体外杀伤比例如图7所示。5) Killing ratio calculation formula: 100% × (blank hole reading - experimental hole reading) / blank hole reading, the in vitro killing ratio of SC-CTL cells to human leukemia cell line NALM6 is shown in FIG. 7 .
实施例5、SP-CTL细胞对人非小细胞性肺癌H460的体内杀伤效应Example 5: In vivo killing effect of SP-CTL cells on human non-small cell lung cancer H460
1)将1×105个H460-GL细胞皮下移植入免疫缺陷小鼠NOD/SCID IL2rg-/-体内;1) 1×10 5 H460-GL cells were subcutaneously transplanted into immunodeficient mice NOD/SCID IL2rg-/-;
2)10天后,在H460-GL荷瘤小鼠中注射SP-CTL GFP阳性细胞数为4×106的SP-CTL病毒感染混合细胞,15天后第二次注射4×106的SP-CTL病毒感染混合细胞,对照组为和上述相同操作的注射GFP T细胞的荷瘤小鼠,每个实验组设置三个重复;2) 10 days after injection of the H460-GL tumor bearing mice SP-CTL GFP positive cells to SP-CTL 4 × 10 6 virus infected cells were mixed, 15 days after the second injection of 4 × 10 6 SP-CTL The virus infects the mixed cells, and the control group is the tumor-bearing mice injected with GFP T cells in the same operation as above, and three replicates are set for each experimental group;
3)于肿瘤移植后,第10、14、17、20、23、26、29、32、35天量取H460肿瘤块体积,结果如图8所示。3) After tumor transplantation, the volume of H460 tumor mass was measured on days 10, 14, 17, 20, 23, 26, 29, 32, and 35, and the results are shown in Fig. 8.
通过以上检测结果可知,本发明的SP-CTL细胞对表达相应配体的各种各样的肿瘤具有显著的体内、外杀伤效应。From the above test results, it is understood that the SP-CTL cells of the present invention have significant in vivo and exogenous killing effects on various tumors expressing the corresponding ligands.
实施例6、SP-CTL细胞的安全性实验Example 6, safety experiment of SP-CTL cells
1)在不表达配体PD-L1的肺腺癌细胞系A549和慢性髓系白血病细胞系K562中转导表达荧光素酶(Luciferace),构建A549-GL、K562-GL报告细胞系;1) Transduction of luciferase (Luciferace) in lung adenocarcinoma cell line A549 and chronic myeloid leukemia cell line K562 which do not express ligand PD-L1, and construction of A549-GL, K562-GL reporter cell line;
2)将SP-CTL GFP阳性细胞分别与A549-GL、K562-GL细胞以细胞数1∶2比例混合共培养(对照组为加GFP T细胞,每组设置三个复孔);2) SP-CTL GFP-positive cells were co-cultured with A549-GL and K562-GL cells at a ratio of 1:2 (the control group was GFP T cells, and three replicate wells were set in each group);
3)18小时后,轻轻用移液器吸除一半体积的培养基上清,加入等体积的荧光素酶 底物(浓度:300μg/ml),混匀;3) After 18 hours, gently pipette half of the medium supernatant and add an equal volume of luciferase Substrate (concentration: 300 μg/ml), mix;
4)通过多功能酶标仪检测SP-CTL细胞对A549-GL、K562-GL细胞的体外杀伤比例,其结果分别如图9、10所示。4) The in vitro killing ratio of SP-CTL cells to A549-GL and K562-GL cells was detected by a multi-function microplate reader, and the results are shown in Figures 9 and 10, respectively.
由以上检测结果可知,SP-CTL细胞仅对表达PD-L1的靶细胞具有特异性杀伤作用,对不表达PD-L1的细胞没有杀伤作用,具有稳定的生物安全性。It can be seen from the above results that SP-CTL cells have a specific killing effect only on target cells expressing PD-L1, no killing effect on cells not expressing PD-L1, and have stable biosafety.
实施例7、SP-CTL细胞体外识别杀伤肺癌、前列腺癌细胞Example 7, SP-CTL cells were identified in vitro to kill lung cancer and prostate cancer cells
1)通过流式细胞技术检测肺癌细胞系(H1299、H460、H23、A549)、前列腺癌细胞系(PC3)细胞中配体PD-L1的表达情况,其结果图11所示;1) The expression of ligand PD-L1 in lung cancer cell lines (H1299, H460, H23, A549) and prostate cancer cell line (PC3) cells was detected by flow cytometry, and the results are shown in Fig. 11;
2)将SP-CTL GFP阳性细胞分别与用荧光素酶GL标记的H1299、H460、H23、A549、PC3细胞(分别简称H1299-GL、H460-GL、H23-GL、A549-GL、PC3-GL)以细胞数1∶1、1∶2、1∶4、1∶8、1∶16比例混合共培养(对照组为加GFP T细胞,每组设置三个复孔);2) SP-CTL GFP-positive cells were separately labeled with H1299, H460, H23, A549, and PC3 cells labeled with luciferase GL (H1299-GL, H460-GL, H23-GL, A549-GL, PC3-GL, respectively) The cells were co-cultured in a ratio of 1:1, 1:2, 1:4, 1:8, 1:16 (the control group was GFP T cells, and three replicate wells were set in each group);
3)18小时后,轻轻用移液器吸除一半体积的培养基上清,加入等体积的荧光素酶底物(浓度:300μg/ml),混匀;3) After 18 hours, gently pipette a half volume of the medium supernatant, add an equal volume of luciferase substrate (concentration: 300 μg / ml), and mix;
4)通过多功能酶标仪检测SP-CTL细胞对H1299-GL、H460-GL、H23-GL、A549-GL、PC3-GL细胞的体外杀伤比例,其结果如图12所示。4) The in vitro killing ratio of SP-CTL cells to H1299-GL, H460-GL, H23-GL, A549-GL and PC3-GL cells was detected by a multi-function microplate reader. The results are shown in Fig. 12.
由以上检测结果可知,SP-CTL细胞可特异性识别、杀伤表达PD-L1的靶细胞(肺癌细胞、前列腺癌细胞等)。From the above test results, SP-CTL cells can specifically recognize and kill target cells (lung cancer cells, prostate cancer cells, etc.) expressing PD-L1.
实施例8、SP-CTL细胞体内识别杀伤原代肺癌细胞Example 8, SP-CTL cells recognize killer primary lung cancer cells in vivo
1)用剪刀将原代肺癌组织样本剪成直径为3mm的肿瘤组织小块,皮下移植入免疫缺陷小鼠NOD/SCID IL2rg-/-体内,构建肺癌小鼠模型;1) Cut the primary lung cancer tissue sample into small pieces of tumor tissue with a diameter of 3 mm, and transplant it into the immunodeficient mouse NOD/SCID IL2rg-/- to construct a lung cancer mouse model.
2)20天后,在肺癌小鼠模型中注射SP-CTL GFP阳性细胞数为4×106的SP-CTL病毒感染混合细胞,25天第二次注射4×106的SP-CTL病毒感染混合细胞,对照组为 注射GFP T细胞的荷瘤小鼠,每个实验组设置三个重复;2) 20 days after injection of lung cancer in a mouse model of SP-CTL of GFP positive cells of 4 × 10 6 SP-CTL infected cells were mixed, 25 days of the second injection of 4 × 10 6 SP-CTL mixed infection The cells and the control group were tumor-bearing mice injected with GFP T cells, and three replicates were set for each experimental group;
3)于肿瘤移植后第20、24、28、20、23、26、29、32、35天,量取肺癌皮下肿瘤组织块的体积;在不同时间点,各实验组肿瘤组织块的平均体积结果如图13-A所示。3) On the 20th, 24th, 28th, 20th, 23rd, 26th, 29th, 32th and 35th day after tumor transplantation, the volume of subcutaneous tumor tissue in lung cancer was measured; the average volume of tumor tissue blocks in each experimental group at different time points The result is shown in Figure 13-A.
4)于肿瘤移植后第44天,从肺癌小鼠模型中获取肿瘤组织块;每个实验组三个重复的肿瘤组织块的尺寸比对图片如图13-B所示。4) Tumor tissue blocks were obtained from the mouse model of lung cancer on the 44th day after tumor transplantation; the size alignment of the three replicated tumor tissue blocks in each experimental group is shown in Fig. 13-B.
通过以上检测结果可知,本发明的SP-CTL细胞在原代肿瘤异质性、免疫抑制微环境等情况下,仍可在体内有效清除原代肺癌细胞。From the above test results, it can be seen that the SP-CTL cells of the present invention can effectively eliminate primary lung cancer cells in vivo in the case of primary tumor heterogeneity and immunosuppressive microenvironment.
实施例9、SP-CTL细胞体内识别杀伤原代胃癌细胞Example 9. SP-CTL cells were identified in vivo to kill primary gastric cancer cells
1)用剪刀将原代胃癌组织样本剪成直径为3mm的肿瘤组织小块,皮下移植入免疫缺陷小鼠NOD/SCID IL2rg-/-体内,构建胃癌小鼠模型;1) Cut the primary gastric cancer tissue sample into small pieces of tumor tissue with a diameter of 3 mm, and transplant it into the immunodeficient mouse NOD/SCID IL2rg-/- to construct a gastric cancer mouse model.
2)20天后,在胃癌小鼠模型中注射SP-CTL GFP阳性细胞数为4×106的SP-CTL病毒感染混合细胞,25天第二次注射4×106的SP-CTL病毒感染混合细胞,对照组为注射GFP T细胞的荷瘤小鼠,每个实验组设置三个重复;2) After 20 days, a SP-CTL virus-infected mixed cell with SP-CTL GFP positive cells of 4×10 6 was injected into a mouse model of gastric cancer, and a second injection of 4×10 6 SP-CTL virus infection was mixed for 25 days. The cells and the control group were tumor-bearing mice injected with GFP T cells, and three replicates were set for each experimental group;
3)于肿瘤移植后第44天,从胃癌小鼠模型中获取肿瘤组织块;每个实验组三个重复的肿瘤组织块的尺寸比对图片如图14-A所示,每个实验组的肿瘤组织块的平均重量结果如图14-B所示。3) On the 44th day after tumor transplantation, tumor tissue blocks were obtained from the mouse model of gastric cancer; the size comparison of the three replicated tumor tissue blocks in each experimental group is shown in Figure 14-A, for each experimental group. The mean weight results for the tumor tissue mass are shown in Figure 14-B.
通过以上检测结果可知,本发明的SP-CTL细胞在原代肿瘤异质性、免疫抑制微环境等情况下,仍可在体内有效清除原代胃癌细胞。From the above test results, it can be seen that the SP-CTL cells of the present invention can effectively eliminate primary gastric cancer cells in vivo in the case of primary tumor heterogeneity and immunosuppressive microenvironment.
实施例10、SP-CTL细胞体内识别杀伤原代骨肉瘤细胞Example 10, SP-CTL cells in vivo to identify primary osteosarcoma cells
1)用剪刀将原代骨肉瘤组织样本剪成直径为3mm的肿瘤组织小块,皮下移植入免疫缺陷小鼠NOD/SCID IL2rg-/-体内,构建骨肉瘤小鼠模型;1) Cut the original osteosarcoma tissue sample into small pieces of tumor tissue with a diameter of 3 mm, and transplant it into the immunodeficient mouse NOD/SCID IL2rg-/- to construct a mouse model of osteosarcoma.
2)12天后,在骨肉瘤小鼠模型中注射SP-CTL GFP阳性细胞数为4×106的SP-CTL病毒感染混合细胞,14天第二次注射4×106的SP-CTL病毒感染混合细胞,对照组为 注射GFP T细胞的荷瘤小鼠,每个实验组设置六个重复;2) 12 days after injection of osteosarcoma tumor mouse model SP-CTL GFP positive cells to SP-CTL 4 × 10 6 virus infected cells were mixed, 14 second injection of 4 × 10 6 SP-CTL infected The cells were mixed, and the control group was tumor-bearing mice injected with GFP T cells, and six replicates were set for each experimental group;
3)于肿瘤移植后第38天,从骨肉瘤小鼠模型中获取肿瘤组织块;每个实验组六个重复的肿瘤组织块的尺寸比对图片如图15-A所示,每个实验组的肿瘤组织块的平均重量结果如图15-B所示。3) On the 38th day after tumor transplantation, tumor tissue blocks were obtained from the osteosarcoma mouse model; the size comparison images of six replicate tumor tissue blocks in each experimental group are shown in Figure 15-A, and each experimental group The average weight results for the tumor tissue mass are shown in Figure 15-B.
通过以上检测结果可知,本发明的SP-CTL细胞在原代肿瘤异质性、免疫抑制微环境等情况下,仍可在体内有效清除原代骨肉瘤细胞。From the above test results, it can be seen that the SP-CTL cells of the present invention can effectively remove primary osteosarcoma cells in vivo in the case of primary tumor heterogeneity and immunosuppressive microenvironment.
实施例11、SC-CTL细胞体外特异性识别杀伤非霍金森淋巴瘤RL细胞Example 11, SC-CTL cells specifically recognize killer non-Hawking's lymphoma RL cells in vitro
1)将SC-CTL GFP阳性细胞与非霍金森淋巴瘤RL细胞(细胞系均用荧光素酶GL标记)以细胞数2∶1、1∶1、0.5∶1、0.25∶1、1∶16比例混合共培养(对照组为加GFP T细胞,每组设置三个复孔);1) SC-CTL GFP-positive cells and non-Hawkinson's lymphoma RL cells (cell lines are labeled with luciferase GL) at a cell number of 2:1, 1:1, 0.5:1, 0.25:1, 1:16 Proportional mixed co-culture (control group was added GFP T cells, each set of three duplicate wells);
2)18小时后,轻轻用移液器吸除一半体积的培养基上清,加入等体积的荧光素酶底物(浓度:300μg/ml),混匀;2) After 18 hours, gently pipette a half volume of the medium supernatant, add an equal volume of luciferase substrate (concentration: 300 μg / ml), and mix;
3)通过多功能酶标仪检测SC-CTL细胞对RL-GL细胞的体外杀伤比例,其结果如图16所示。3) The in vitro killing ratio of SC-CTL cells to RL-GL cells was detected by a multi-function microplate reader, and the results are shown in Fig. 16.
由以上检测结果可知,SC-CTL细胞可在体外特异性识别和杀伤非霍金森淋巴瘤细胞。From the above test results, SC-CTL cells can specifically recognize and kill non-Hawkinson lymphoma cells in vitro.
实施例12、SC-CTL细胞体内特异性识别杀伤非霍金森淋巴瘤RL细胞Example 12, SC-CTL cells specifically recognize killer non-Hawking's lymphoma RL cells in vivo
1)将1×105个RL-GL细胞皮下移植入免疫缺陷小鼠NOD/SCID IL2rg-/-体内,构建RL-GL荷瘤小鼠模型;1) 1×10 5 RL-GL cells were subcutaneously transplanted into immunodeficient mouse NOD/SCID IL2rg-/- to construct a RL-GL tumor-bearing mouse model;
2)2天后,在RL-GL荷瘤小鼠中注射SC-CTL GFP阳性细胞数为1×105的SC-CTL病毒感染混合细胞,对照组为注射GFP T细胞的荷瘤小鼠,每个实验组设置三个重复;2) Two days later, RL-GL tumor-bearing mice were injected with SC-CTL virus-positive cells with a population of 1 × 10 5 SC-CTL virus-infected mixed cells, and the control group was tumor-bearing mice injected with GFP T cells. Three experimental groups set up three repetitions;
3)于肿瘤移植后第28天,从RL-GL荷瘤小鼠小鼠模型中获取肿瘤组织块;每个实验组两个重复的肿瘤组织块的尺寸比对图片如图17-A所示,每个实验组的肿瘤组织 块的平均重量结果如图17-B所示。3) On the 28th day after tumor transplantation, tumor tissue blocks were obtained from the mouse model of RL-GL tumor-bearing mice; the size comparison of two repeated tumor tissue blocks in each experimental group is shown in Figure 17-A. Tumor tissue of each experimental group The average weight result of the block is shown in Figure 17-B.
通过以上检测结果可知,本发明的SC-CTL细胞可在体内有效清除非霍金森淋巴瘤细胞。From the above test results, it is known that the SC-CTL cells of the present invention can effectively eliminate non-Hawsonson lymphoma cells in vivo.
实施例13、mSC-CTL细胞体内特异性识别杀伤原代骨肉瘤细胞Example 13, mSC-CTL cells specifically recognize killer primary osteosarcoma cells in vivo
通过分子克隆手段,用小鼠CTLA4胞外段+跨膜区(核苷酸序列和氨基酸序列分别如SEQ ID NO.5、SEQ ID NO.6所示)替换(h)SC-CTL的胞外段+跨膜区,获得mSC-CTL。本实施例目的在于评估mSC-CTL细胞在体内对原代骨肉瘤细胞的特异性识别杀伤作用。具体程序如下:Substitution of mouse extracellular domain + transmembrane region of CTLA4 (nucleotide sequence and amino acid sequence as shown in SEQ ID NO. 5, SEQ ID NO. 6) by molecular cloning means (h) extracellularization of SC-CTL Segment + transmembrane region, mSC-CTL was obtained. The purpose of this example was to evaluate the specific recognition and killing effect of mSC-CTL cells on primary osteosarcoma cells in vivo. The specific procedures are as follows:
1)用剪刀将原代骨肉瘤组织样本剪成直径为3mm的肿瘤组织小块,皮下移植入免疫缺陷小鼠NOD/SCID IL2rg-/-体内(每只受体小鼠移植2小块),构建骨肉瘤小鼠模型;1) Cut the original osteosarcoma tissue sample into small pieces of tumor tissue with a diameter of 3 mm and subcutaneously transplant it into the immunodeficient mouse NOD/SCID IL2rg-/- (2 small pieces per recipient mouse). Constructing a mouse model of osteosarcoma;
2)20天后,在骨肉瘤小鼠模型中注射mSC-CTL GFP阳性细胞数为1×105的mSC-CTL病毒感染混合细胞,对照组为注射GFP T细胞的荷瘤小鼠,每个实验组设置三个重复;2) 20 days later, in the osteosarcoma mouse model, mSC-CTL GFP-positive cells were injected with mixed cells of m××10 5 mSC-CTL virus, and the control group was tumor-bearing mice injected with GFP T cells. The group sets three repetitions;
3)于肿瘤移植44天,从骨肉瘤小鼠模型中获取肿瘤组织块;每个实验组的肿瘤组织块的尺寸比对图片如图18-A所示,每个实验组的肿瘤组织块的平均重量结果如图18-B所示。3) Tumor tissue blocks were obtained from the osteosarcoma mouse model 44 days after tumor transplantation; the size of the tumor tissue blocks in each experimental group was shown in Fig. 18-A, and the tumor tissue blocks of each experimental group were The average weight results are shown in Figure 18-B.
通过以上检测结果可知,本发明的mSC-CTL细胞在原代肿瘤异质性、免疫抑制微环境等情况下,仍可在体内有效清除原代骨肉瘤细胞。From the above test results, it can be seen that the mSC-CTL cells of the present invention can effectively eliminate primary osteosarcoma cells in vivo in the case of primary tumor heterogeneity and immunosuppressive microenvironment.
实施例14、mSC-CTL和SP-CTL联合应用体内识别杀伤肿瘤细胞Example 14, in combination with mSC-CTL and SP-CTL to identify killing tumor cells in vivo
通过分子克隆手段,用小鼠CTLA4胞外段+跨膜区(核苷酸序列和氨基酸序列分别如SEQ.NO 5、6所示)替换(h)SC-CTL的胞外段+跨膜区,获得mSC-CTL;本实施例目的在于评估SP-CTL和mSC-CTL的联合应用对肿瘤细胞的清除作用,评估 SC-CTL对小鼠CD80+/CD86+肿瘤辅助细胞(非肿瘤细胞)的识别杀伤作用,并评估该作用在肿瘤清除中的影响。具体程序如下:Substitution of mouse extracellular domain + transmembrane region (nucleotide sequence and amino acid sequence as shown in SEQ. NO 5, 6) by mouse cloning means (h) extracellular domain + transmembrane region of SC-CTL , obtaining mSC-CTL; the purpose of this example is to evaluate the scavenging effect of SP-CTL and mSC-CTL on tumor cells, and evaluate SC-CTL recognizes the killing effect of mouse CD80+/CD86+ tumor helper cells (non-tumor cells) and evaluates the effect of this effect on tumor clearance. The specific procedures are as follows:
1)用剪刀将原代肺癌组织样本剪成直径为3mm的肿瘤组织小块,皮下移植入免疫缺陷小鼠NOD/SCID IL2rg-/-体内,构建肺癌小鼠模型;1) Cut the primary lung cancer tissue sample into small pieces of tumor tissue with a diameter of 3 mm, and transplant it into the immunodeficient mouse NOD/SCID IL2rg-/- to construct a lung cancer mouse model.
2)18天后,将肺癌小鼠模型分为四组,分别注射SP-CTL GFP阳性细胞数为2×105的SP-CTL病毒感染混合细胞(SP-CTL组),mSC-CTL GFP阳性细胞数为2×105的SP-CTL病毒感染混合细胞(mSC-CTL组),以及混合的2×105SP-CTL GFP阳性细胞和2×105mSC-CTL GFP阳性细胞(SP-CTL+mSC-CTL联合应用组),对照组为注射GFP T细胞的荷瘤小鼠,每个实验组设置三个重复;2) After 18 days, the lung cancer mouse model was divided into four groups, and SP-CTL virus-infected mixed cells (SP-CTL group) with SP-CTL GFP positive cells of 2×10 5 were injected, and mSC-CTL GFP-positive cells were injected. 2 × 10 5 SP-CTL virus-infected mixed cells (mSC-CTL group), and mixed 2×10 5 SP-CTL GFP-positive cells and 2×10 5 mSC-CTL GFP-positive cells (SP-CTL+) mSC-CTL combined application group), the control group was tumor-bearing mice injected with GFP T cells, and each experiment group was set up with three repetitions;
3)于肿瘤移植后第18、21、24、27、30、33、36天,量取肺癌皮下肿瘤块体积;在不同时间点,各实验组肿瘤组织块的平均体积结果如图19-A所示。3) The volume of subcutaneous tumor mass of lung cancer was measured on the 18th, 21st, 24th, 27th, 30th, 33rd and 36th day after tumor transplantation. At different time points, the average volume of tumor tissue blocks in each experimental group is shown in Figure 19-A. Shown.
4)于肿瘤移植后第38天,从肺癌小鼠模型中获取肿瘤组织块,每个实验组的肿瘤组织块的平均重量结果如图19-B所示;该结果显示:本发明的SP-CTL联合mSC-CTL细胞可在体内有效且协同清除原代肺癌细胞,且效果比单独使用SP-CTL或mSC-CTL细胞更为显著。4) On the 38th day after tumor transplantation, tumor tissue blocks were obtained from the mouse model of lung cancer, and the average weight results of the tumor tissue blocks of each experimental group are shown in Fig. 19-B; the results show: SP- of the present invention CTL combined with mSC-CTL cells can effectively and synergistically eliminate primary lung cancer cells in vivo, and the effect is more significant than SP-CTL or mSC-CTL cells alone.
5)取SP-CTL、mSC-CTL、SP-CTL+mSC-CTL联合应用组的肿瘤组织块,研磨、消化为单细胞,通过流式细胞技术检测肿瘤组织块中小鼠的CD80、CD86、MDSC(CD11b+Ly6G+)细胞的比例,其结果如图20-A、20-B、20-C所示。5) Take the tumor tissue blocks of SP-CTL, mSC-CTL, SP-CTL+mSC-CTL combined application group, grind and digest into single cells, and detect CD80, CD86, MDSC of mice in tumor tissue block by flow cytometry. The ratio of (CD11b+Ly6G+) cells, and the results are shown in Figures 20-A, 20-B, and 20-C.
由以上检测结果可得出如下结论:mSC-CTL不仅可识别杀伤肿瘤细胞,还可识别杀伤CD80+或/和CD86+细胞(除肿瘤细胞外的肿瘤辅助细胞),而且mSC-CTL对CD80+/CD86+非肿瘤细胞的杀伤作用可有效抑制肿瘤生长。mSC-CTL联合SP-CTL可同时识别杀伤肿瘤细胞和CD80+/CD86+肿瘤辅助性细胞,阻断CD80+/CD86+肿瘤辅助性细胞对肿瘤细胞生长、迁移、耐药、免疫逃逸等的作用,达到更好的肿瘤清除效 果。From the above test results, the following conclusions can be drawn: mSC-CTL can not only identify killing tumor cells, but also recognize CD80+ or/and CD86+ cells (except tumor helper cells other than tumor cells), and mSC-CTL for CD80+/CD86+ The killing effect of tumor cells can effectively inhibit tumor growth. mSC-CTL combined with SP-CTL can simultaneously identify killer tumor cells and CD80+/CD86+ tumor-assisted cells, and block the effects of CD80+/CD86+ tumor-assisted cells on tumor cell growth, migration, drug resistance, immune escape, etc. Tumor elimination effect fruit.
实施例15、白血病和淋巴瘤细胞高表达Lag3配体MHC II(图21)Example 15. High expression of Lag3 ligand MHC II in leukemia and lymphoma cells (Fig. 21)
通过流式细胞技术检测人B细胞白血病系Nalm6、B细胞淋巴瘤细胞系Raji和RL细胞表面分子人HLA-DR(HLA-DR为MHC II分子关键组分)的表达情况,发现以上三种细胞系均特异性高表达人HLA-DR基因,即特异性高表达MHC II分子(Lag3配体),表明人B细胞白血病和B细胞淋巴瘤的肿瘤细胞是SL-CTL的潜在靶点细胞。Flow cytometry was used to detect the expression of human B cell leukemia cell line Nalm6, B cell lymphoma cell line Raji and RL cell surface molecule human HLA-DR (HLA-DR is a key component of MHC II molecule). The lines all specifically express human HLA-DR gene, which specifically expresses MHC II molecules (Lag3 ligand), indicating that human B cell leukemia and B cell lymphoma tumor cells are potential target cells of SL-CTL.
实施例16、SL-CTL可特异性识别杀伤表达Lag3配体的肿瘤细胞(图22、23)Example 16. SL-CTL specifically recognizes tumor cells that secrete Lag3 ligand (Figures 22, 23)
1)通过(实施例1)慢病毒包装pWPXLd-SL-CTL-2A-EGFP质粒并感染人原代T细胞,获得SL-CTL细胞。1) SL-CTL cells were obtained by packaging (p. 1) lentivirus pWPXLd-SL-CTL-2A-EGFP plasmid and infecting human primary T cells.
2)将培养扩增后的SL-CTL细胞与靶细胞Nalm6-GL细胞在96孔板中以不同E(Effector)∶T(Target)比2∶1、1∶1、1∶2、1∶4、1∶8混合,共培养18小时。2) Incubating the expanded SL-CTL cells with the target cells Nalm6-GL cells in a 96-well plate at a different E(Effector):T(Target) ratio of 2:1, 1:1, 1:2, 1: 4. Mix 1:8 and co-culture for 18 hours.
3)轻轻用移液器吸除一半体积的培养基上清,加入等体积的荧光素酶底物(浓度:300μg/ml),混匀;3) gently pipette half of the medium supernatant, add an equal volume of luciferase substrate (concentration: 300μg / ml), and mix;
4)通过多功能酶标仪检测SL-CTL细胞对A549-GL、K562-GL细胞的体外杀伤比例,其结果分别如图22所示。4) The in vitro killing ratio of SL-CTL cells to A549-GL and K562-GL cells was detected by a multi-function microplate reader, and the results are shown in Fig. 22, respectively.
5)SL-CTL细胞(空白对照组GFP-T)与Nalm6-GL细胞以1∶1混合共培养18小时后,通过ELISA检测培养基上清中的白细胞介素2(IL-2)和干扰素γ(IFNγ)的水平,如图23所示。结果显示,与野生T细胞(GFP-T组)相比,SL-CTL细胞能够更高效地杀伤Nalm6靶细胞,而且SL-CTL细胞可分泌更多的炎症因子IL-2和IFNγ。5) SL-CTL cells (blank control group GFP-T) and Nalm6-GL cells were co-cultured for 1:1 with 1:1, and the interleukin-2 (IL-2) and interference in the culture supernatant were detected by ELISA. The level of gamma (IFNγ) is shown in Figure 23. The results showed that SL-CTL cells were able to kill Nalm6 target cells more efficiently than wild T cells (GFP-T group), and SL-CTL cells secreted more inflammatory factors IL-2 and IFNγ.
实施例17、体内的肿瘤细胞可上调自身TIM3配体Galectin9的表达(图24、25)Example 17, tumor cells in vivo can up-regulate the expression of its own TIM3 ligand Galectin9 (Figures 24, 25).
1)通过流式细胞技术检测A549细胞株不表达Galectin9;1) Detection of A549 cell line by flow cytometry does not express Galectin9;
2)将A549细胞移植入免疫缺陷小鼠(如NOD/SCID IL2rg-/-)体内,21天后, 取小鼠肿瘤块,研磨裂解成单细胞悬液,检测Galectin9阳性细胞比例,如图24所示;2) A549 cells were transplanted into immunodeficient mice (such as NOD/SCID IL2rg-/-), 21 days later, The tumor block of the mouse was taken, ground and lysed into a single cell suspension, and the proportion of Galectin9-positive cells was detected, as shown in FIG. 24;
3)将#1、#2、#3三例病人来源的肺癌样本细胞/组织移植入免疫缺陷小鼠体内,60天后,取小鼠体内人体肺癌肿瘤组织块,研磨裂解为单细胞悬液,通过流式细胞技术检测肿瘤组织块中Galectin9阳性细胞比例,如图25所示。3) Three cells of human lung cancer samples from #1, #2, and #3 were transplanted into immunodeficient mice. After 60 days, the human lung cancer tumor tissue was taken from the mouse and ground into a single cell suspension. The proportion of Galectin9 positive cells in tumor tissue blocks was detected by flow cytometry as shown in FIG.
4)本实施例数据表明,体内的肿瘤细胞(更接近于病人体内的肿瘤状态)相对于体外培养的肿瘤细胞系,特异性上调Galectin9(Tim的配体)表达,表明体内状态(亦包含病人体内状态的肿瘤细胞)的肿瘤细胞是ST-CTL细胞的潜在特异性靶细胞。4) The data in this example show that tumor cells in vivo (closer to the tumor state in the patient) specifically up-regulate the expression of Galectin9 (Tim ligand) relative to the in vitro cultured tumor cell line, indicating in vivo status (including patients) Tumor cells of tumor cells in an in vivo state are potential specific target cells of ST-CTL cells.
实施例18、ST-CTL细胞可在体内特异性识别和杀伤表达TIM3配体的肿瘤细胞(图26)Example 18, ST-CTL cells specifically recognize and kill tumor cells expressing TIM3 ligand in vivo (Fig. 26)
1)用剪刀将原代肺癌组织样本#2剪成直径为3mm的肿瘤组织小块,皮下移植入免疫缺陷小鼠NOD/SCID IL2rg-/-体内,构建肺癌小鼠模型;1) Cut the primary lung cancer tissue sample #2 into small pieces of tumor tissue with a diameter of 3 mm, and transplant it into the immunodeficient mouse NOD/SCID IL2rg-/- to construct a lung cancer mouse model.
2)20天后,在肺癌小鼠模型中注射ST-CTL GFP阳性细胞数为4×106的ST-CTL病毒感染混合细胞,25天第二次注射GFP阳性细胞数4×106的SP-CTL病毒感染混合细胞,对照组为注射GFP T细胞的荷瘤小鼠,空白(NC)组为无处理(不注射任何T细胞)的荷瘤小鼠,每个实验组设置三个重复;2) 20 days after injection of lung cancer in a mouse model ST-CTL of GFP positive cells of 4 × 10 6 ST-CTL infected cells were mixed, a second injection 25 days the number of GFP-positive cells in the SP- 4 × 10 6 The CTL virus infects mixed cells, the control group is tumor-bearing mice injected with GFP T cells, and the blank (NC) group is tumor-free mice without treatment (no T cells are injected), and three replicates are set for each experimental group;
3)于肿瘤移植后第45天,从三个组实验肺癌小鼠模型中获取肿瘤组织块;并称量肿瘤组织块重量,结果如图26所示。3) On the 45th day after tumor transplantation, tumor tissue blocks were obtained from three groups of experimental lung cancer mouse models; the tumor tissue mass was weighed, and the results are shown in Fig. 26.
4)结果发现ST-CTL细胞可在体内抑制肿瘤细胞的生长。4) It was found that ST-CTL cells can inhibit the growth of tumor cells in vivo.
申请人声明,本发明通过上述实施例来说明本发明的产品、用途及其使用方式,但本发明并不局限于上述详细用途和使用方式,即不意味着本发明必须依赖上述详细用途和使用方式才能实施。所属技术领域的技术人员应该明了,对本发明的任何改进,对本发明产品各原料的等效替换及辅助成分的添加、具体方式的选择等,均落在本发明的保护范围和公开范围之内。 The Applicant declares that the present invention describes the products, uses, and uses thereof of the present invention by the above embodiments, but the present invention is not limited to the above detailed uses and modes of use, that is, does not mean that the present invention must rely on the above detailed uses and uses. The way can be implemented. It should be apparent to those skilled in the art that any modifications of the present invention, equivalent substitution of the various materials of the products of the present invention, addition of auxiliary components, selection of specific means, and the like, are all within the scope of the present invention.
本文所涉及基因序列的列表List of gene sequences involved in this article
SEQ ID NO.1 PD-1信号肽+胞外段+跨膜区-CD28-TLR1/2-CD3ζSEQ ID NO.1 PD-1 signal peptide + extracellular domain + transmembrane region - CD28-TLR1/2-CD3ζ
Figure PCTCN2017088594-appb-000001
Figure PCTCN2017088594-appb-000001
SEQ ID NO.2 CTLA-4信号肽+胞外段+跨膜区-CD28胞内段-TLR1/2胞内段-CD3ζSEQ ID NO.2 CTLA-4 signal peptide + extracellular domain + transmembrane region - CD28 intracellular segment - TLR1/2 intracellular segment - CD3
Figure PCTCN2017088594-appb-000002
Figure PCTCN2017088594-appb-000002
Figure PCTCN2017088594-appb-000003
Figure PCTCN2017088594-appb-000003
SEQ ID NO.3 TIM3信号肽+胞外段+跨膜区-4-1BB胞内段-TLR2胞内段-CD3ζSEQ ID NO.3 TIM3 signal peptide + extracellular domain + transmembrane region - 4-1BB intracellular segment - TLR2 intracellular segment - CD3
Figure PCTCN2017088594-appb-000004
Figure PCTCN2017088594-appb-000004
SEQ ID NO.4 LAG3信号肽+胞外段+跨膜区-4-1BB胞内段-TLR2胞内段-CD3ζSEQ ID NO.4 LAG3 signal peptide + extracellular domain + transmembrane region - 4-1BB intracellular segment - TLR2 intracellular segment - CD3
Figure PCTCN2017088594-appb-000005
Figure PCTCN2017088594-appb-000005
Figure PCTCN2017088594-appb-000006
Figure PCTCN2017088594-appb-000006
SEQ IDNO.5 mCTLA4胞外段+跨膜区SEQ ID NO.5 mCTLA4 extracellular domain + transmembrane region
Figure PCTCN2017088594-appb-000007
Figure PCTCN2017088594-appb-000007
SEQ.ID NO.6 mCTLA4胞外段+跨膜区SEQ.ID NO.6 mCTLA4 extracellular domain + transmembrane region
Figure PCTCN2017088594-appb-000008
Figure PCTCN2017088594-appb-000008

Claims (10)

  1. 一种嵌合的免疫抑制检查点受体分子,其包含胞外结构域、跨膜结构域和胞内结构域,其中,所述胞外结构域和任选地跨膜结构域为免疫抑制检查点受体分子的相应结构域或基于该结构域进行的基因改造,所述胞内结构域为一个或多个免疫激活信号结构域。A chimeric immunosuppressive checkpoint receptor molecule comprising an extracellular domain, a transmembrane domain and an intracellular domain, wherein the extracellular domain and optionally a transmembrane domain are immunosuppressive assays A corresponding domain of a point receptor molecule or a genetic modification based on the domain, the intracellular domain being one or more immune activation signal domains.
  2. 根据权利要求1所述的免疫抑制检查点受体分子,其特征在于,所述免疫抑制检查点受体分子选自PD-1、CTLA-4、LAG3、TIM3、A2AR、B7H3、B7H4、BTLA、IDO、KIR、CD160、2B4(CD244)和VISTA(C10orf54);The immunosuppressive checkpoint receptor molecule according to claim 1, wherein the immunosuppressive checkpoint receptor molecule is selected from the group consisting of PD-1, CTLA-4, LAG3, TIM3, A2AR, B7H3, B7H4, BTLA, IDO, KIR, CD160, 2B4 (CD244) and VISTA (C10orf54);
    优选地,所述免疫检查点受体分子为PD-1、CTLA-4、LAG3或TIM3。Preferably, the immune checkpoint receptor molecule is PD-1, CTLA-4, LAG3 or TIM3.
  3. 根据权利要求1或2所述的免疫抑制检查点受体分子,其特征在于,所述胞内结构域包含免疫共刺激信号胞内域组合和任选的信号肽;The immunosuppressive checkpoint receptor molecule according to claim 1 or 2, wherein the intracellular domain comprises an immunostimulation signal intracellular domain combination and an optional signal peptide;
    优选地,所述免疫共刺激信号组合包含TLR1和/或TLR2信号结构域;Preferably, the immune costimulatory signal combination comprises a TLR1 and/or TLR2 signal domain;
    进一步优选地,所述胞内结构域为TLR1和/或TLR2与41BB和/或CD28等信号结构域的组合;Further preferably, the intracellular domain is a combination of TLR1 and/or TLR2 and a signal domain such as 41BB and/or CD28;
    优选地,任选的信号肽为免疫抑制检查点受体分子相对应的信号肽。Preferably, the optional signal peptide is a signal peptide corresponding to an immunosuppressive checkpoint receptor molecule.
  4. 根据权利要求3所述的免疫抑制检查点受体分子,其特征在于,所述胞内结构域还包括CD3ζ胞内结构域;The immunosuppressive checkpoint receptor molecule according to claim 3, wherein the intracellular domain further comprises a CD3 sputum intracellular domain;
    优选地,所述TLR1和/或TLR2信号结构域和任选地41BB和/或CD28信号结构域配置在CD3ζ胞内结构域的N末端侧。Preferably, the TLR1 and/or TLR2 signal domain and optionally the 41BB and/or CD28 signal domain are arranged on the N-terminal side of the CD3 cell intracellular domain.
  5. 编码如权利要求1-4任一项所述的嵌合的免疫抑制检查点受体分子的核酸。A nucleic acid encoding a chimeric immunosuppressive checkpoint receptor molecule of any of claims 1-4.
  6. 一种核酸构建体,其包含如权利要求5所述的核酸,以及与之可操作连接、可指导所述嵌合的免疫抑制检查点受体分子在宿主细胞中表达的一个或多个控制序列。A nucleic acid construct comprising the nucleic acid of claim 5, and one or more control sequences operably linked thereto for directing expression of said chimeric immunosuppressive checkpoint receptor molecule in a host cell .
  7. 一种表达载体,其包含如权利要求6所述的核酸构建体;优选地,所述表达载体为慢病毒表达载体。 An expression vector comprising the nucleic acid construct of claim 6; preferably, the expression vector is a lentiviral expression vector.
  8. 一种转化的细胞,其包含如权利要求5所述的核酸,或者其中转化了如权利要求6所述的核酸构建体或如权利要求7所述的表达载体;A transformed cell comprising the nucleic acid of claim 5, or wherein the nucleic acid construct of claim 6 or the expression vector of claim 7 is transformed;
    优选地,所述转化的细胞为免疫细胞,优选为免疫效应细胞,进一步优选为T细胞、B细胞或NK细胞。Preferably, the transformed cell is an immune cell, preferably an immune effector cell, further preferably a T cell, a B cell or an NK cell.
  9. 制备如权利要求8所述的转化的细胞的方法,其包括:向细胞基因组中引入如权利要求5所述的核酸,或者转化如权利要求6所述的核酸构建体或如权利要求7所述的表达载体的步骤。A method of producing the transformed cell of claim 8, comprising: introducing the nucleic acid of claim 5 into the genome of the cell, or transforming the nucleic acid construct of claim 6 or the method of claim 7 The step of the expression vector.
  10. 如权利要求1-4任一项所述的嵌合的免疫抑制检查点受体分子、如权利要求5所述的核酸、如权利要求6所述的核酸构建体、如权利要求7所述的表达载体或如权利要求8所述的转化的细胞在制备与免疫抑制检查点受体分子对应的配体、优选PD-L1、B7.1、B7.2、Galectin-9、MHC II的表达相关的疾病的药物中的用途;A chimeric immunosuppressive checkpoint receptor molecule according to any one of claims 1 to 4, a nucleic acid according to claim 5, a nucleic acid construct according to claim 6, or a method according to claim 7. The expression vector or the transformed cell of claim 8 is associated with the expression of a ligand corresponding to an immunosuppressive checkpoint receptor molecule, preferably PD-L1, B7.1, B7.2, Galectin-9, MHC II Use of the drug for the disease;
    优选地,所述疾病为表达免疫抑制检查点受体分子对应的配体的肿瘤、优选表达PD-L1、B7.1、B7.2、Galectin-9、MHC II的肿瘤;Preferably, the disease is a tumor expressing a ligand corresponding to an immunosuppressive checkpoint receptor molecule, preferably a tumor expressing PD-L1, B7.1, B7.2, Galectin-9, MHC II;
    进一步优选地,所述表达免疫抑制检查点受体分子对应的配体的肿瘤、优选表达PD-L1、B7.1、B7.2、Galectin-9、MHC II的肿瘤选自肺癌、白血病、乳腺癌、前列腺癌、胰腺癌、肝癌、黑色素瘤和非黑色素瘤皮肤癌。 Further preferably, the tumor expressing the ligand corresponding to the immunosuppressive checkpoint receptor molecule, preferably the tumor expressing PD-L1, B7.1, B7.2, Galectin-9, MHC II is selected from the group consisting of lung cancer, leukemia, and mammary gland Cancer, prostate cancer, pancreatic cancer, liver cancer, melanoma, and non-melanoma skin cancer.
PCT/CN2017/088594 2016-06-20 2017-06-16 Molecule, cell expressing the same, and preparation method and use thereof WO2017219916A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202110620458.7A CN113512126B (en) 2016-06-20 2017-06-16 Molecule, cell expressing same and preparation method and application thereof
CN201780000466.3A CN107624119B (en) 2016-06-20 2017-06-16 Molecule, cell expressing same, preparation method and application thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201610443073.7 2016-06-20
CN201610443073.7A CN107522786A (en) 2016-06-20 2016-06-20 A kind of molecule, the cell for expressing it and its production and use

Publications (1)

Publication Number Publication Date
WO2017219916A1 true WO2017219916A1 (en) 2017-12-28

Family

ID=60734431

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/088594 WO2017219916A1 (en) 2016-06-20 2017-06-16 Molecule, cell expressing the same, and preparation method and use thereof

Country Status (2)

Country Link
CN (3) CN107522786A (en)
WO (1) WO2017219916A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11708423B2 (en) 2017-09-26 2023-07-25 Cero Therapeutics, Inc. Chimeric engulfment receptor molecules and methods of use

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112226412B (en) * 2020-10-12 2023-05-02 汤朝阳 T cell expressing immunosuppressive checkpoint receptor molecule and application thereof
CN114081941A (en) * 2021-11-19 2022-02-25 上海奢旭企业管理中心(有限合伙) Preparation method of B7-H4 protein and application of B7-H4 protein in preparation of medicine for resisting excessive immune response or resisting cytokine storm
CN116731205B (en) * 2023-05-19 2024-02-27 皖南医学院第一附属医院(皖南医学院弋矶山医院) Immunopotentiating fusion protein for expressing TGF beta RII and TIM3 extracellular region and application thereof

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014031687A1 (en) * 2012-08-20 2014-02-27 Jensen, Michael Method and compositions for cellular immunotherapy
CN103965361A (en) * 2013-02-06 2014-08-06 上海细胞治疗工程技术研究中心有限公司 Single chimeric converter for T-cell signal and application thereof
WO2015095895A1 (en) * 2013-12-20 2015-06-25 Fred Hutchinson Cancer Research Center Tagged chimeric effector molecules and receptors thereof
WO2015112626A1 (en) * 2014-01-21 2015-07-30 June Carl H Enhanced antigen presenting ability of car t cells by co-introduction of costimulatory molecules
CN104829733A (en) * 2015-05-25 2015-08-12 广州科锐特生物科技有限公司 Chimeric antigen receptor with stable antigen binding units, method for preparing chimeric antigen receptor and application thereof
CN105153315A (en) * 2015-10-09 2015-12-16 重庆倍思益生物科技有限公司 Chimeric receptor combining immunosuppression receptor and tumor antigen receptor and application of chimeric receptor
CN105331585A (en) * 2015-11-13 2016-02-17 科济生物医药(上海)有限公司 Chimeric antigen receptor-modified immunologic effector cell with PD-L1 blocking agent
CN105601752A (en) * 2016-02-01 2016-05-25 长沙郝怡雅医药科技有限公司 Multi-gene recombination chimeric antigen receptor molecule

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP7372728B2 (en) * 2014-10-31 2023-11-01 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Methods and compositions related to modified T cells
CN104673897B (en) * 2015-01-26 2017-09-29 江苏先声药业有限公司 A kind of method of the biological activity of the measure pathway inhibitors of PD 1

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014031687A1 (en) * 2012-08-20 2014-02-27 Jensen, Michael Method and compositions for cellular immunotherapy
CN103965361A (en) * 2013-02-06 2014-08-06 上海细胞治疗工程技术研究中心有限公司 Single chimeric converter for T-cell signal and application thereof
WO2015095895A1 (en) * 2013-12-20 2015-06-25 Fred Hutchinson Cancer Research Center Tagged chimeric effector molecules and receptors thereof
WO2015112626A1 (en) * 2014-01-21 2015-07-30 June Carl H Enhanced antigen presenting ability of car t cells by co-introduction of costimulatory molecules
CN104829733A (en) * 2015-05-25 2015-08-12 广州科锐特生物科技有限公司 Chimeric antigen receptor with stable antigen binding units, method for preparing chimeric antigen receptor and application thereof
CN105153315A (en) * 2015-10-09 2015-12-16 重庆倍思益生物科技有限公司 Chimeric receptor combining immunosuppression receptor and tumor antigen receptor and application of chimeric receptor
CN105331585A (en) * 2015-11-13 2016-02-17 科济生物医药(上海)有限公司 Chimeric antigen receptor-modified immunologic effector cell with PD-L1 blocking agent
CN105601752A (en) * 2016-02-01 2016-05-25 长沙郝怡雅医药科技有限公司 Multi-gene recombination chimeric antigen receptor molecule

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11708423B2 (en) 2017-09-26 2023-07-25 Cero Therapeutics, Inc. Chimeric engulfment receptor molecules and methods of use

Also Published As

Publication number Publication date
CN113512126B (en) 2023-05-05
CN113512126A (en) 2021-10-19
CN107624119A (en) 2018-01-23
CN107522786A (en) 2017-12-29
CN107624119B (en) 2021-07-09

Similar Documents

Publication Publication Date Title
US20210253726A1 (en) Car expression vector and car-expressing t cells
EP3680338A1 (en) Genetically engineered t cell and application thereof
CN109803983B (en) Specific chimeric antigen receptor T cell targeting NKG2DL, preparation method and application thereof
WO2019061562A1 (en) Nucleic acid molecule which enhances antitumor activity of t cells
Ptáčková et al. A new approach to CAR T-cell gene engineering and cultivation using piggyBac transposon in the presence of IL-4, IL-7 and IL-21
JP2019510503A (en) Chimeric antigen receptor T cell composition
WO2017219916A1 (en) Molecule, cell expressing the same, and preparation method and use thereof
AU2019275479B2 (en) Chimeric antigen receptors with modified linker domains and uses thereof
CN114656569B (en) Multispecific chimeric receptor comprising NKG2D domains and methods of use thereof
WO2015166056A1 (en) Cs1 specific multi-chain chimeric antigen receptor
JP2019512251A (en) PD-1 and 4-1BB fusion proteins
Tal et al. An NCR1-based chimeric receptor endows T-cells with multiple anti-tumor specificities
CN112533957A (en) MUC 16-specific chimeric antigen receptor and uses thereof
AU2021219233A1 (en) Immune cells overexpressing cell signaling regulatory factor introduced from outside and use thereof
WO2020118634A1 (en) Immune effector cell targeting gpc3 and application thereof
WO2019097083A1 (en) Modified k562 cell
WO2022007796A1 (en) Immune cell co-expressing il-21 and hrcd16 chimeric receptor, and application thereof
Huye et al. Designing T cells for cancer immunotherapy
WO2023125860A1 (en) Preparation technique for universal car-t cell, and application of universal car-t cell
EP4353253A1 (en) Purification of tcr-modified t cells using tcr-specific car-nk cells
Dowell Studies of human T cell costimulation: potential for the immunotherapy of cancer

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17814654

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 20/05/2019)

122 Ep: pct application non-entry in european phase

Ref document number: 17814654

Country of ref document: EP

Kind code of ref document: A1